1
|
Allerdice MEJ, Shooter SL, Galletti MFBM, Hecht JA, Karpathy SE, Paddock CD. Molecular identification and antibiotic clearance of Mycoplasma arginini and Mycoplasma orale from cell cultures infected with Rickettsia or Ehrlichia species. Microbiol Spectr 2025:e0174324. [PMID: 39817787 DOI: 10.1128/spectrum.01743-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 12/02/2024] [Indexed: 01/18/2025] Open
Abstract
Mycoplasma (Class: Mollicutes) contamination in cell cultures is a universal concern for research laboratories. Some estimates report contamination in up to 35% of continuous cell lines. Various commercial antibiotic treatments can successfully decontaminate clean cell lines in vitro; however, in vitro decontamination of bacterial cultures remains challenging. Intracellular bacteria like those in the genera Rickettsia and Ehrlichia require cell culture for primary isolation and propagation and are thus vulnerable to contamination with mycoplasmas. Some analyses have reported successful antibiotic clearance of contaminating mycoplasmas in Rickettsia cultures; however, many of these studies do not identify the contaminating mycoplasma species and often include only a few isolates. To our knowledge, there are no published studies reporting decontamination of mycoplasmas from Ehrlichia cultures. In this study, we developed a specific multiplex assay to identify two of the most common mycoplasma culture contaminants, Mycoplasma arginini and Mycoplasma orale, in cell cultures infected with Rickettsia or Ehrlichia species. We further describe the successful in vitro decontamination of M. arginini, M. orale, and co-contaminations with both mycoplasmas from multiple Rickettsia and Ehrlichia cultures using daptomycin and clindamycin.IMPORTANCEMycoplasma contamination is a frequent problem in bacterial cell culture. These prolific organisms thrive in the extracellular environment in vitro and can persist in cell lines indefinitely without treatment. Historically, mycoplasma-contaminated Rickettsia cultures were cleared of contaminants by inoculating laboratory mice and re-isolating mycoplasma-free Rickettsia from brain endothelial cells. However, this method requires the sacrifice of live animals and is not always effective. Mycoplasma clearance via mouse inoculation requires a patent infection of murine central nervous system endothelial cells, which may not occur with some mildly pathogenic or nonpathogenic rickettsial species. In vitro antibiotic treatment represents an alternate method to eliminate contaminating mycoplasmas from rickettsial cultures. This method requires minimal adjustment of laboratories that already maintain rickettsial cultures and is not dependent on the use of laboratory animals. As such, the comprehensive strategy for Mycoplasma arginini and Mycoplasma orale elimination presented here can improve laboratory efficiency for in vitro research with intracellular bacteria.
Collapse
Affiliation(s)
- Michelle E J Allerdice
- Division of Vector-borne Diseases, Centers for Disease Control and Prevention, U.S. Department of Health and Human Services, Atlanta, Georgia, USA
| | - Savannah L Shooter
- Rickettsial Zoonoses Branch, Centers for Disease Control and Prevention, U.S. Department of Health and Human Services, Atlanta, Georgia, USA
| | - Maria F B M Galletti
- Rickettsial Zoonoses Branch, Centers for Disease Control and Prevention, U.S. Department of Health and Human Services, Atlanta, Georgia, USA
| | - Joy A Hecht
- Rickettsial Zoonoses Branch, Centers for Disease Control and Prevention, U.S. Department of Health and Human Services, Atlanta, Georgia, USA
| | - Sandor E Karpathy
- Rickettsial Zoonoses Branch, Centers for Disease Control and Prevention, U.S. Department of Health and Human Services, Atlanta, Georgia, USA
| | - Christopher D Paddock
- Rickettsial Zoonoses Branch, Centers for Disease Control and Prevention, U.S. Department of Health and Human Services, Atlanta, Georgia, USA
| |
Collapse
|
2
|
Ferradas C, Salvatierra G, Payahuanca D, Contreras W, López-Pérez AM, Hangawatte TA, León D, Ghersi BM, Gamboa R, Villanueva KM, Pinedo-Cancino V, Pesapane R, Salmón-Mulanovich G, Lescano AG, Foley J. Spotted fever group rickettsiae in black rats, pets, and humans in Zungarococha community, A rural area in the surroundings of Iquitos, Peru. Ticks Tick Borne Dis 2025; 16:102436. [PMID: 39799873 DOI: 10.1016/j.ttbdis.2024.102436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/23/2024] [Accepted: 12/31/2024] [Indexed: 01/15/2025]
Abstract
Rickettsiae are a family of ectoparasite-borne bacteria that can produce high morbidity and mortality among humans. There are scarce data on rickettsial ecology in rural areas of the Peruvian Amazon basin, where seroprevalence has not been determined, and the identities of animals acting as reservoirs of these bacteria are not known. We conducted a cross-sectional study in Zungarococha (between 2019 and 2021), a rural community located approximately 20 km away from Iquitos city. Blood samples were collected from humans (175), dogs (123), and cats (12). Blood samples and tissues were collected from black rats (84). Finally, we collected fleas from dogs and cats (222), ticks from dogs (91), and mites from black rats (32). Blood samples from humans, dogs, cats, and black rats were analyzed by indirect immunofluorescence assays (IFA) to detect IgG antibodies against rickettsias. We screened ectoparasites and black rat tissues by real-time-PCR (qPCR). Positive ectoparasites were further assessed by PCR and DNA amplicon sequencing. Non-parametric tests were used to evaluate factors associated with being seropositive among human adults. IgG seroprevalences were 38.3 %, 58.5 %, 16.7 % and 48.1 % among humans, dogs, cats, and rats, respectively. Among humans, only male gender was statistically associated with having IgG antibodies against Rickettsia spp. (p-value=0.049, chi-square test). Different ectoparasites were identified, including Ctenocephalides felis from cats and dogs, Rhipicephalus sanguineus s.l. from dogs, and Laelaps nuttalli from black rats. Rhipicephalus sanguineus s.l. (2/91 ticks) and Ct. felis (53/56 fleas and 55/55 flea pools) were qPCR-positive for Rickettsia spp. Recovered genetic material from 53 Ct. felis was sequenced and all were identified as Rickettsia asembonensis. All tissue samples from black rats were negative by qPCR. Humans, dogs, cats, and black rats are exposed to spotted fever group rickettsiae in rural areas surrounding Iquitos. As reported in urban areas, R. asembonensis is the main Rickettsia species circulating in rural areas surrounding Iquitos and Ct. felis appears to be the main vector.
Collapse
Affiliation(s)
- Cusi Ferradas
- Emerge, Emerging Diseases and Climate Change Research Unit, School of Public Health and Administration, Universidad Peruana Cayetano Heredia (UPCH), Lima Peru; Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, CA USA
| | - Guillermo Salvatierra
- Department of Health Sciences, School of Veterinary Medicine, Universidad Peruana de Ciencias Aplicadas (UPC), Lima Peru
| | - David Payahuanca
- Emerge, Emerging Diseases and Climate Change Research Unit, School of Public Health and Administration, Universidad Peruana Cayetano Heredia (UPCH), Lima Peru
| | - Winnie Contreras
- Emerge, Emerging Diseases and Climate Change Research Unit, School of Public Health and Administration, Universidad Peruana Cayetano Heredia (UPCH), Lima Peru
| | - Andrés M López-Pérez
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, CA USA; Red de Biología y Conservación de Vertebrados, Instituto de Ecología, A.C, Xalapa Mexico
| | - Therangika A Hangawatte
- School of Environment and Natural Resources, College of Food, Agricultural, and Environmental Sciences, The Ohio State University, OH USA
| | - Diana León
- Emerge, Emerging Diseases and Climate Change Research Unit, School of Public Health and Administration, Universidad Peruana Cayetano Heredia (UPCH), Lima Peru
| | - Bruno M Ghersi
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, MA USA
| | - Ricardo Gamboa
- Center for Global Health, Universidad Peruana Cayetano Heredia (UPCH), Tumbes Peru
| | | | - Viviana Pinedo-Cancino
- Laboratorio de Investigación en Productos Naturales Antiparasitarios de la Amazonía, Centro de Investigación de Recursos Naturales de la Amazonía, Univesidad Nacional de la Amazonía Peruana (UNAP), Iquitos Peru; Facultad de Medicina Humana. Universidad Nacional de la Amazonia Peruana (UNAP), Iquitos Peru
| | - Risa Pesapane
- School of Environment and Natural Resources, College of Food, Agricultural, and Environmental Sciences, The Ohio State University, OH USA; Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, OH USA
| | | | - Andrés G Lescano
- Emerge, Emerging Diseases and Climate Change Research Unit, School of Public Health and Administration, Universidad Peruana Cayetano Heredia (UPCH), Lima Peru; Clima, Latin American Center of Excellence for Climate Change and Health, Universidad Peruana Cayetano Heredia (UPCH), Lima Peru
| | - Janet Foley
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, CA USA.
| |
Collapse
|
3
|
Kim JK, Sapkota A, Roh T, Jo EK. The intricate interactions between inflammasomes and bacterial pathogens: Roles, mechanisms, and therapeutic potentials. Pharmacol Ther 2025; 265:108756. [PMID: 39581503 DOI: 10.1016/j.pharmthera.2024.108756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 10/06/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024]
Abstract
Inflammasomes are intracellular multiprotein complexes that consist of a sensor, an adaptor, and a caspase enzyme to cleave interleukin (IL)-1β and IL-18 into their mature forms. In addition, caspase-1 and -11 activation results in the cleavage of gasdermin D to form pores, thereby inducing pyroptosis. Activation of the inflammasome and pyroptosis promotes host defense against pathogens, whereas dysregulation of the inflammasome can result in various pathologies. Inflammasomes exhibit versatile microbial signal detection, directly or indirectly, through cellular processes, such as ion fluctuations, reactive oxygen species generation, and the disruption of intracellular organelle function; however, bacteria have adaptive strategies to manipulate the inflammasome by altering microbe-associated molecular patterns, intercepting innate pathways with secreted effectors, and attenuating inflammatory and cell death responses. In this review, we summarize recent advances in the diverse roles of the inflammasome during bacterial infections and discuss how bacteria exploit inflammasome pathways to establish infections or persistence. In addition, we highlight the therapeutic potential of harnessing bacterial immune subversion strategies against acute and chronic bacterial infections. A more comprehensive understanding of the significance of inflammasomes in immunity and their intricate roles in the battle between bacterial pathogens and hosts will lead to the development of innovative strategies to address emerging threats posed by the expansion of drug-resistant bacterial infections.
Collapse
Affiliation(s)
- Jin Kyung Kim
- Department of Microbiology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Asmita Sapkota
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea; Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Taylor Roh
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea; Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea; Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea.
| |
Collapse
|
4
|
Roxas PB, Cruz J, Horelka NR, Burgos C, Radwanski J, Baires F, Sierra-Hoffman M, Hesse H, Madril AC. Typhus group Rickettsia community-acquired bacterial central nervous system infections: We must think outside the box! J Neurol Sci 2024; 466:123281. [PMID: 39447222 DOI: 10.1016/j.jns.2024.123281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/24/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024]
Abstract
Typhus group rickettsiosis (TGR), caused by Rickettsia typhi and Rickettsia prowazekii, are globally distributed vector-borne diseases with increasing cases. Diagnosis is usually clinical, confirmed by seroconversion of IgG antibodies. Human infection occurs in diverse geographic areas with some developing CNS infection characterized by fever, headache, meningismus, and/or focal signs - usually beyond the first week of initial symptomatology. Seizures and other CNS manifestations have been observed. When untreated, infection may result in neurologic sequelae and even death. This study presents a systematic review of all documented cases of Rickettsia typhi meningoencephalitis published since 2015 with the addition of five cases of TGR in South Coastal Texas, USA. This review followed the guidelines outlined in PRISMA. A schematic explanation of the pathophysiology is offered. CSF may present with high opening pressure, mild to moderate pleocytosis, mildly elevated protein levels, and low csf/serum glucose ratio, or normal findings. Meningeal enhancement, intracranial hypertension, and focal abnormalities have been described in imaging studies, but can be normal. Treatment with doxycycline leads to prompt resolution of symptoms. Failure to initiate early empiric treatment can lead to serious consequences. The study recommends routine testing for TGR in patients from endemic areas with classical symptoms when other diagnoses are inconclusive or in cases with atypical presentations. The authors advocate for incorporating empiric treatment for murine typhus into community-acquired bacterial meningitis guidelines in endemic areas; and stress the importance of enhancing laboratory diagnostic capabilities in public health entities world-wide. Further studies of community acquired mengingoencephalitis caused by TGR are highly encouraged.
Collapse
Affiliation(s)
- Pauline B Roxas
- Family Medicine Residency, Detar Healthcare System - Texas A&M University School of Medicine, Victoria, TX, USA
| | - Justice Cruz
- Victoria College, Department of Science, Victoria, TX, USA.
| | | | - Cesar Burgos
- Universidad Nacional Autónoma de Honduras, Instituto Nacional Cardiopulmonar El Tórax, Tegucigalpa, Honduras
| | | | - Fernando Baires
- Universidad Nacional Autónoma de Honduras, Tegucigalpa, Honduras
| | - Miguel Sierra-Hoffman
- El Campo Memorial Hospital, El Campo, Texas, USA; Clinical Professor of Texas A&M, Rural Health Medicine Residency Program, Victoria, Texas, USA
| | - Heike Hesse
- Instituto de Investigaciones One Health, Universidad Tecnológica Centroamericana, Tegucigalpa, Honduras.
| | - Amy C Madril
- Department of Hospital Medicine, El Campo Memorial Hospital, El Campo, TX, USA
| |
Collapse
|
5
|
Rattanakomol P, Khongwichit S, Poovorawan Y. Flea-Borne Rickettsioses and Scrub Typhus in Patients with Suspected Arbovirus Infection in Bangkok, Thailand. Vector Borne Zoonotic Dis 2024; 24:649-655. [PMID: 38946645 DOI: 10.1089/vbz.2024.0030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2024] Open
Abstract
Background: In urban Thailand, arboviral infections dominate diagnoses of acute undifferentiated fevers (AUFs) owing to their well-defined epidemiology and characteristic clinical presentations. However, rickettsial diseases, also endemic in this setting, remain under-recognized owing to challenges in early detection. Objective: This study aimed to identify potential rickettsial infections among patients with AUF in Bangkok and vicinity utilizing leftover nucleic acid extracted from serum samples from patients initially suspected of but negative for arbovirus infections. Materials and Methods: A total of 609 nucleic acid samples were screened for rickettsial bacteria using real-time PCR, targeting the 17-kDa common antigen gene of Rickettsia spp. and the 47-kDa gene of Orientia tsutsugamushi. Results: Nine samples were positive for Rickettsia spp. and two were positive for O. tsutsugamushi. DNA sequence and phylogenetic analyses based on partial 17-kDa antigen and citrate synthase (gltA) genes identified the Rickettsia-positive samples as R. typhi in eight cases and R. felis in one case. Analysis of the 56-kDa type-specific antigen gene identified the two O. tsutsugamushi isolates as Gilliam-related genotypes. Although rickettsial diseases typically present with mild symptoms, two patients with R. typhi infection (murine typhus) developed respiratory distress syndrome, highlighting the potential for rare but serious complications. Conclusion: This study underscores the critical importance of differential diagnosis and prompt, effective intervention to prevent complications in suspected cases.
Collapse
Affiliation(s)
- Patthaya Rattanakomol
- Department of Pediatrics, Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Sarawut Khongwichit
- Department of Pediatrics, Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Yong Poovorawan
- Department of Pediatrics, Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
6
|
Baharani A, Reddy P RR. Treatment Outcomes of Presumed Rickettsial Retinitis: Evidence from OCTA Based Quantitative Analysis. Ocul Immunol Inflamm 2024; 32:1197-1204. [PMID: 37141535 DOI: 10.1080/09273948.2023.2206490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/16/2023] [Accepted: 04/19/2023] [Indexed: 05/06/2023]
Abstract
AIM To quantify retinal ischemia following presumed rickettsial retinitis (RR). To compare outcomes between initial treatment with Doxycycline (Group 1) versus steroids (Group 2). METHODS Retrospective analysis of patients with presumed RR was done. ImageJ software was used to obtain %area of ischemia on swept-source optical coherence tomography angiography (SS-OCTA). RESULTS Eleven eyes of 8 patients belonged to Group1 and 6 eyes of 3 patients belonged to Group 2. The BCVA improved from logMAR 0.8 ∓ 0.7 to logMAR 0.06 ∓ 0.08 (p < 0.002) and central foveal thickness (CFT) changed from 479μ ∓ 341.3μ to 163.5μ ∓ 20.5μ (p < 0.005) after a median of 5 weeks in Group 1. In Group 2, BCVA improved from logMAR 1.03 ∓ 0.05 to logMAR 0.23 ∓ 0.23 (p < 0.004) and CFT changed from 286.5μ ∓ 158.8μ to 177.5μ ∓ 25.9μ (>0.05) following a mean of 11 weeks. Mean %area of ischemia was 4.6 ∓ 1.5 in Group 1 and 13.9 ∓ 4.1 in Group 2. CONCLUSION Analysis of flow deficit on SS-OCTA confirms that treatment with Doxycycline in presumed RR results in less ischemia and quicker recovery than initial treatment with steroids.
Collapse
|
7
|
Perumalsamy N, Sharma R, Subramanian M, Nagarajan SA. Hard Ticks as Vectors: The Emerging Threat of Tick-Borne Diseases in India. Pathogens 2024; 13:556. [PMID: 39057783 PMCID: PMC11279560 DOI: 10.3390/pathogens13070556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 07/28/2024] Open
Abstract
Hard ticks (Ixodidae) play a critical role in transmitting various tick-borne diseases (TBDs), posing significant global threats to human and animal health. Climatic factors influence the abundance, diversity, and vectorial capacity of tick vectors. It is imperative to have a comprehensive understanding of hard ticks, pathogens, eco-epidemiology, and the impact of climatic changes on the transmission dynamics of TBDs. The distribution and life cycle patterns of hard ticks are influenced by diverse ecological factors that, in turn, can be impacted by changes in climate, leading to the expansion of the tick vector's range and geographical distribution. Vector competence, a pivotal aspect of vectorial capacity, involves the tick's ability to acquire, maintain, and transmit pathogens. Hard ticks, by efficiently feeding on diverse hosts and manipulating their immunity through their saliva, emerge as competent vectors for various pathogens, such as viruses, parasites and bacteria. This ability significantly influences the success of pathogen transmission. Further exploration of genetic diversity, population structure, and hybrid tick vectors is crucial, as they play a substantial role in influencing vector competence and complicating the dynamics of TBDs. This comprehensive review deals with important TBDs in India and delves into a profound understanding of hard ticks as vectors, their biology, and the factors influencing their vector competence. Given that TBDs continue to pose a substantial threat to global health, the review emphasizes the urgency of investigating tick control strategies and advancing vaccine development. Special attention is given to the pivotal role of population genetics in comprehending the genetic diversity of tick populations and providing essential insights into their adaptability to environmental changes.
Collapse
Affiliation(s)
| | | | | | - Shriram Ananganallur Nagarajan
- Division of Vector Biology and Control, Indian Council of Medical Research—Vector Control Research Centre (ICMR-VCRC), Puducherry 605006, India; (N.P.); (R.S.); (M.S.)
| |
Collapse
|
8
|
Pustijanac E, Buršić M, Millotti G, Paliaga P, Iveša N, Cvek M. Tick-Borne Bacterial Diseases in Europe: Threats to public health. Eur J Clin Microbiol Infect Dis 2024; 43:1261-1295. [PMID: 38676855 DOI: 10.1007/s10096-024-04836-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 04/22/2024] [Indexed: 04/29/2024]
Abstract
BACKGROUND Tick-borne diseases, caused by bacterial pathogens, pose a growing threat to public health in Europe. This paper provides an overview of the historical context of the discovery of the most impactful pathogens transmitted by ticks, including Borrelia burgdorferi sensu lato, Rickettsia spp., Anaplasma spp., Francisella spp., Ehrlichia spp., and Neoehrlichia mikurensis. Understanding the historical context of their discovery provides insight into the evolution of our understanding of these pathogens. METHODS AND RESULTS Systematic investigation of the prevalence and transmission dynamics of these bacterial pathogens is provided, highlighting the intricate relationships among ticks, host organisms, and the environment. Epidemiology is explored, providing an in-depth analysis of clinical features associated with infections. Diagnostic methodologies undergo critical examination, with a spotlight on technological advancements that enhance detection capabilities. Additionally, the paper discusses available treatment options, addressing existing therapeutic strategies and considering future aspects. CONCLUSIONS By integrating various pieces of information on these bacterial species, the paper aims to provide a comprehensive resource for researchers and healthcare professionals addressing the impact of bacterial tick-borne diseases in Europe. This review underscores the importance of understanding the complex details influencing bacterial prevalence and transmission dynamics to better combat these emerging public health threats.
Collapse
Affiliation(s)
- Emina Pustijanac
- Faculty of Natural Sciences, Juraj Dobrila University of Pula, Zagrebačka 30, 52100, Pula, Croatia.
| | - Moira Buršić
- Faculty of Natural Sciences, Juraj Dobrila University of Pula, Zagrebačka 30, 52100, Pula, Croatia
| | - Gioconda Millotti
- Faculty of Natural Sciences, Juraj Dobrila University of Pula, Zagrebačka 30, 52100, Pula, Croatia
| | - Paolo Paliaga
- Faculty of Natural Sciences, Juraj Dobrila University of Pula, Zagrebačka 30, 52100, Pula, Croatia
| | - Neven Iveša
- Faculty of Natural Sciences, Juraj Dobrila University of Pula, Zagrebačka 30, 52100, Pula, Croatia
| | - Maja Cvek
- Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000, Rijeka, Croatia
- Teaching Institute of Public Health of the Region of Istria, Nazorova 23, 52100, Pula, Croatia
| |
Collapse
|
9
|
Brito-Lorán CB, Araiza-Rodríguez A, Garcés-Ayala F, Contreras-Pérez CU, Montes-Colima NA, López-Martínez I, Hernandez-Cortez C, Castro-Escarpulli G, Ramírez-González JE. Analysis of Rocky Mountain spotted fever cases in Northern Mexico reveals genetic variability of Rickettsia rickettsii and the different distribution of genotypes. Int Microbiol 2024; 27:689-695. [PMID: 37646898 DOI: 10.1007/s10123-023-00424-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 08/16/2023] [Accepted: 08/23/2023] [Indexed: 09/01/2023]
Abstract
Rickettsioses have been reported in parts of Mexico since the last century, with Rocky Mountain spotted fever (RMSF) being one of the most prevalent in northern states. Unfortunately, fatality rates for RMSF in Mexico are higher than in other countries, like the USA. The reason for this difference in fatality rates is currently unknown and could be associated with a genotype of the bacterium, but no comparative molecular typing has been conducted in Mexico to date. The purpose of this study was to analyze 47 RMSF samples with different outcomes from several states in northern Mexico to know the genetic variability of Rickettsia rickettsii, as well as to reconstruct its phylogeny, for which the following intergenic regions were sequenced: RR0155-rpmB, cspA-ksgA, RR1240-tlc5, and Spo0J-abc T1, as well as the following partial genes: ompA, ompB, and gltA. We identified 8 genotypes with different distribution and prevalence among the states analyzed, as well as a different association with case outcome; these genotypes were clustered in 2 clades and 5 lineages were revealed, some of them probably exclusive from Mexico.
Collapse
Affiliation(s)
- Carina Berenice Brito-Lorán
- Laboratorio de Investigación Clínica y Ambiental, Departamento de Bioquímica Microbiana, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), Colonia Santo Tomás, Miguel Hidalgo, 011340, México City, México
- Instituto de Diagnóstico y Referencia Epidemiológicos (InDRE) "Dr. Manuel Martínez Báez", Secretaría de Salud, Francisco de P. Miranda 177, Lomas de Plateros, Álvaro Obregón, 01480, Mexico City, México
| | - Adnan Araiza-Rodríguez
- Instituto de Diagnóstico y Referencia Epidemiológicos (InDRE) "Dr. Manuel Martínez Báez", Secretaría de Salud, Francisco de P. Miranda 177, Lomas de Plateros, Álvaro Obregón, 01480, Mexico City, México
| | - Fabiola Garcés-Ayala
- Instituto de Diagnóstico y Referencia Epidemiológicos (InDRE) "Dr. Manuel Martínez Báez", Secretaría de Salud, Francisco de P. Miranda 177, Lomas de Plateros, Álvaro Obregón, 01480, Mexico City, México
| | - Cudberto U Contreras-Pérez
- Instituto de Diagnóstico y Referencia Epidemiológicos (InDRE) "Dr. Manuel Martínez Báez", Secretaría de Salud, Francisco de P. Miranda 177, Lomas de Plateros, Álvaro Obregón, 01480, Mexico City, México
| | - Norma Angélica Montes-Colima
- Instituto de Diagnóstico y Referencia Epidemiológicos (InDRE) "Dr. Manuel Martínez Báez", Secretaría de Salud, Francisco de P. Miranda 177, Lomas de Plateros, Álvaro Obregón, 01480, Mexico City, México
| | - Irma López-Martínez
- Instituto de Diagnóstico y Referencia Epidemiológicos (InDRE) "Dr. Manuel Martínez Báez", Secretaría de Salud, Francisco de P. Miranda 177, Lomas de Plateros, Álvaro Obregón, 01480, Mexico City, México
| | - Cecilia Hernandez-Cortez
- Laboratorio de Investigación Clínica y Ambiental, Departamento de Bioquímica Microbiana, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), Colonia Santo Tomás, Miguel Hidalgo, 011340, México City, México
| | - Graciela Castro-Escarpulli
- Laboratorio de Investigación Clínica y Ambiental, Departamento de Bioquímica Microbiana, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), Colonia Santo Tomás, Miguel Hidalgo, 011340, México City, México.
| | - José Ernesto Ramírez-González
- Instituto de Diagnóstico y Referencia Epidemiológicos (InDRE) "Dr. Manuel Martínez Báez", Secretaría de Salud, Francisco de P. Miranda 177, Lomas de Plateros, Álvaro Obregón, 01480, Mexico City, México.
| |
Collapse
|
10
|
Voss OH, Moin I, Gaytan H, Ullah S, Sadik M, Azad AF, Rahman MS. Pathogenic rickettsiae utilize the phosphatidylserine binding receptor CD300f on macrophages for host invasion and pathogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.10.593542. [PMID: 38766217 PMCID: PMC11100818 DOI: 10.1101/2024.05.10.593542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Some arthropod-borne obligate intracellular rickettsiae are among the most virulent human pathogens. Upon entry, Rickettsia species modulate immune (e.g., macrophages; MΦ) and non-immune cell (e.g., endothelial cells) responses to create a habitable environment for host colonization. In particular, MΦ play a crucial role in either terminating an infection at an early stage or succumbing to bacterial replication and colonization. However, our understanding on how Rickettsia species modulate crucial cellular processes within MΦ, including phagocytosis, and host cell defenses, to establish an intracytosolic replication niche, remain poorly defined. In this study, we describe a previously unappreciated mechanism, in which pathogenic rickettsiae infection is mediated by the phosphatidylserine (PS)-binding receptor, CD300f. We found that CD300f -/- mice but not wild-type (WT) C57BL/6J mice were protected against R. typhi - or R. rickettsii [ Shelia Smith ]-induced fatal rickettsiosis. Adoptative transfer studies further revealed that CD300f-expressing bone marrow-derived macrophages (BMDMΦ) are important mediators to control rickettsiosis in WT mice. Mechanistical analysis, using WT or CD300f -/- BMDMΦ, showed that CD300f facilitates the engulfment of both pathogenic R. typhi and R. rickettsii species, likely via a PS-mediated mechanism. Furthermore, CD300f was involved in the intracytosolic replication of both pathogenic rickettsiae by differentially modulating the anti-inflammatory Interleukin (IL)-10 and anti-rickettsial IL-1α and IL-1β cytokine responses. Collectively, our findings describe a previously unappreciated role for the efferocytic receptor, CD300f, to facilitate engulfment and the intracellular survival of pathogenic rickettsiae within the host. Significance Statement Vector-borne diseases, which are transmitted by hematophagous arthropods, like ticks and fleas, present a perilous threat to public health. In fact, tick- and flea-borne rickettsial diseases are on the rise globally and our current inadequate understanding on how Rickettsia interacts with their mammalian host has significantly impaired the development of effective interventions against pathogenic rickettsial infections. Here, we identified the phosphatidylserine (PS)-receptor, CD300f, as an important mediator of pathogenic rickettsiae infection in vivo and in vitro . Specifically, we showed that CD300f-expressing macrophages facilitate rickettsial infection by differentially modulating anti-inflammatory Interleukin (IL)-10 and anti-rickettsial IL-1α and IL-1β cytokine responses. In sum, our data described CD300f as an important regulator of rickettsial infection and may present a target for therapeutic intervention.
Collapse
|
11
|
Lehman SS, Verhoeve VI, Driscoll TP, Beckmann JF, Gillespie JJ. Metagenome diversity illuminates the origins of pathogen effectors. mBio 2024; 15:e0075923. [PMID: 38564675 PMCID: PMC11077975 DOI: 10.1128/mbio.00759-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 02/12/2024] [Indexed: 04/04/2024] Open
Abstract
Recent metagenome-assembled genome (MAG) analyses have profoundly impacted Rickettsiology systematics. The discovery of basal lineages (novel families Mitibacteraceae and Athabascaceae) with predicted extracellular lifestyles exposed an evolutionary timepoint for the transition to host dependency, which seemingly occurred independent of mitochondrial evolution. Notably, these basal rickettsiae carry the Rickettsiales vir homolog (rvh) type IV secretion system and purportedly use rvh to kill congener microbes rather than parasitize host cells as described for later-evolving rickettsial pathogens. MAG analysis also substantially increased diversity for the genus Rickettsia and delineated a sister lineage (the novel genus Tisiphia) that stands to inform on the emergence of human pathogens from protist and invertebrate endosymbionts. Herein, we probed Rickettsiales MAG and genomic diversity for the distribution of Rickettsia rvh effectors to ascertain their origins. A sparse distribution of most Rickettsia rvh effectors outside of Rickettsiaceae lineages illuminates unique rvh evolution from basal extracellular species and other rickettsial families. Remarkably, nearly every effector was found in multiple divergent forms with variable architectures, indicating profound roles for gene duplication and recombination in shaping effector repertoires in Rickettsia pathogens. Lateral gene transfer plays a prominent role in shaping the rvh effector landscape, as evinced by the discovery of many effectors on plasmids and conjugative transposons, as well as pervasive effector gene exchange between Rickettsia and Legionella species. Our study exemplifies how MAGs can yield insight into pathogen effector origins, particularly how effector architectures might become tailored to the discrete host cell functions of different eukaryotic hosts.IMPORTANCEWhile rickettsioses are deadly vector-borne human diseases, factors distinguishing Rickettsia pathogens from the innumerable bevy of environmental rickettsial endosymbionts remain lacking. Recent metagenome-assembled genome (MAG) studies revealed evolutionary timepoints for rickettsial transitions to host dependency. The rvh type IV secretion system was likely repurposed from congener killing in basal extracellular species to parasitizing host cells in later-evolving pathogens. Our analysis of MAG diversity for over two dozen rvh effectors unearthed their presence in some non-pathogens. However, most effectors were found in multiple divergent forms with variable architectures, indicating gene duplication and recombination-fashioned effector repertoires of Rickettsia pathogens. Lateral gene transfer substantially shaped pathogen effector arsenals, evinced by the discovery of effectors on plasmids and conjugative transposons, as well as pervasive effector gene exchanges between Rickettsia and Legionella species. Our study exemplifies how MAGs yield insight into pathogen effector origins and evolutionary processes tailoring effectors to eukaryotic host cell biology.
Collapse
Affiliation(s)
- Stephanie S. Lehman
- Division of Molecular and Cellular Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Victoria I. Verhoeve
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Timothy P. Driscoll
- Department of Biology, West Virginia University, Morgantown, West Virginia, USA
| | - John F. Beckmann
- Department of Microbiology and Immunology, University of South Alabama, Mobile, Alabama, USA
| | - Joseph J. Gillespie
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
12
|
Kimemia BB, Musila L, Langat S, Odoyo E, Cinkovich S, Abuom D, Yalwala S, Khamadi S, Johnson J, Garges E, Ojwang E, Eyase F. Detection of pathogenic bacteria in ticks from Isiolo and Kwale counties of Kenya using metagenomics. PLoS One 2024; 19:e0296597. [PMID: 38687700 PMCID: PMC11060535 DOI: 10.1371/journal.pone.0296597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 04/09/2024] [Indexed: 05/02/2024] Open
Abstract
Ticks are arachnid ectoparasites that rank second only to mosquitoes in the transmission of human diseases including bacteria responsible for anaplasmosis, ehrlichiosis, spotted fevers, and Lyme disease among other febrile illnesses. Due to the paucity of data on bacteria transmitted by ticks in Kenya, this study undertook a bacterial metagenomic-based characterization of ticks collected from Isiolo, a semi-arid pastoralist County in Eastern Kenya, and Kwale, a coastal County with a monsoon climate in the southern Kenyan border with Tanzania. A total of 2,918 ticks belonging to 3 genera and 10 species were pooled and screened in this study. Tick identification was confirmed through the sequencing of the Cytochrome C Oxidase Subunit 1 (COI) gene. Bacterial 16S rRNA gene PCR amplicons obtained from the above samples were sequenced using the MinION (Oxford Nanopore Technologies) platform. The resulting reads were demultiplexed in Porechop, followed by trimming and filtering in Trimmomatic before clustering using Qiime2-VSearch. A SILVA database pretrained naïve Bayes classifier was used to classify the Operational Taxonomic Units (OTUs) taxonomically. The bacteria of clinical interest detected in pooled tick assays were as follows: Rickettsia spp. 59.43% of pools, Coxiella burnetii 37.88%, Proteus mirabilis 5.08%, Cutibacterium acnes 6.08%, and Corynebacterium ulcerans 2.43%. These bacteria are responsible for spotted fevers, query fever (Q-fever), urinary tract infections, skin and soft tissue infections, eye infections, and diphtheria-like infections in humans, respectively. P. mirabilis, C. acnes, and C. ulcerans were detected only in Isiolo. Additionally, COI sequences allowed for the identification of Rickettsia and Coxiella species to strain levels in some of the pools. Diversity analysis revealed that the tick genera had high levels of Alpha diversity but the differences between the microbiomes of the three tick genera studied were not significant. The detection of C. acnes, commonly associated with human skin flora suggests that the ticks may have contact with humans potentially exposing them to bacterial infections. The findings in this study highlight the need for further investigation into the viability of these bacteria and the competency of ticks to transmit them. Clinicians in these high-risk areas also need to be appraised for them to include Rickettsial diseases and Q-fever as part of their differential diagnosis.
Collapse
Affiliation(s)
- Bryson Brian Kimemia
- Department of Emerging Infectious Diseases, United States Army Medical Research Directorate-Africa (USAMRD-A), Nairobi, Kenya
- Jomo Kenyatta University of Agriculture and Technology (JKUAT), Nairobi, Kenya
| | - Lillian Musila
- Department of Emerging Infectious Diseases, United States Army Medical Research Directorate-Africa (USAMRD-A), Nairobi, Kenya
- Kenya Medical Research Institute (KEMRI), Centre for Microbiology Research, Nairobi, Kenya
| | - Solomon Langat
- Kenya Medical Research Institute (KEMRI), Centre for Virus Research, Nairobi, Kenya
| | - Erick Odoyo
- Department of Emerging Infectious Diseases, United States Army Medical Research Directorate-Africa (USAMRD-A), Nairobi, Kenya
| | - Stephanie Cinkovich
- United States Armed Forces Health Surveillance Division, Global Emerging Infections Surveillance Branch, Silver Spring, Maryland, United States of America
| | - David Abuom
- Department of Emerging Infectious Diseases, United States Army Medical Research Directorate-Africa (USAMRD-A), Nairobi, Kenya
| | - Santos Yalwala
- Department of Emerging Infectious Diseases, United States Army Medical Research Directorate-Africa (USAMRD-A), Nairobi, Kenya
| | - Samoel Khamadi
- Kenya Medical Research Institute (KEMRI), Centre for Virus Research, Nairobi, Kenya
| | - Jaree Johnson
- United States Armed Forces Pest Management Board, Silver Spring, Maryland, United States of America
| | - Eric Garges
- Department of Emerging Infectious Diseases, United States Army Medical Research Directorate-Africa (USAMRD-A), Nairobi, Kenya
| | - Elly Ojwang
- Department of Emerging Infectious Diseases, United States Army Medical Research Directorate-Africa (USAMRD-A), Nairobi, Kenya
| | - Fredrick Eyase
- Department of Emerging Infectious Diseases, United States Army Medical Research Directorate-Africa (USAMRD-A), Nairobi, Kenya
- Kenya Medical Research Institute (KEMRI), Centre for Virus Research, Nairobi, Kenya
| |
Collapse
|
13
|
Fitzsimmons L, Bublitz D, Clark T, Hackstadt T. Rickettsia rickettsii virulence determinants RARP2 and RapL mitigate IFN- β signaling in primary human dermal microvascular endothelial cells. mBio 2024; 15:e0345023. [PMID: 38445878 PMCID: PMC11005427 DOI: 10.1128/mbio.03450-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/13/2024] [Indexed: 03/07/2024] Open
Abstract
We compared the growth characteristics of a virulent Rickettsia rickettsii strain (Sheila Smith) to an attenuated R. rickettsii stain (Iowa) and a non-pathogenic species (R. montanensis) in primary human dermal microvascular endothelial cells (HDMEC). All replicated in Vero cells, however, only the Sheila Smith strain productively replicated in HDMECs. The Iowa strain showed minimal replication over a 24-h period, while R. montanensis lost viability and induced lysis of the HDMECs via a rapid programmed cell death response. Both the virulent and attenuated R. rickettsii strains, but not R. montanensis, induced an interferon-1 response, although the response was of lesser magnitude and delayed in the Sheila Smith strain. IFN-β secretion correlated with increased host cell lysis, and treatment with anti-IFNAR2 antibody decreased lysis from Iowa-infected but not Sheila Smith-infected cells. Both Sheila Smith- and Iowa-infected cells eventually lysed, although the response from Sheila Smith was delayed and showed characteristics of apoptosis. We, therefore, examined whether reconstitution of the Iowa strain with two recently described putative virulence determinants might enhance survival of Iowa within HDMECs. Reconstitution with RARP2, which is inhibitory to anterograde trafficking through the Golgi apparatus, reduced IFN-β secretion but had no effect on cell lysis. RapL, which proteolytically processes surface exposed autotransporters and enhances replication of Iowa in Guinea pigs, suppressed both IFN-β production and host cell lysis. These findings suggest distinct mechanisms by which virulent spotted fever group rickettsiae may enhance intracellular survival and replication.IMPORTANCEWe examined a naturally occurring non-pathogenic rickettsial species, R. montanensis, a laboratory-attenuated R. rickettsii strain (Iowa), and a fully virulent R. rickettsii strain (Sheila Smith) for growth in human dermal microvascular endothelial cells. The two avirulent strains replicated poorly or not at all. Only the virulent Sheila Smith strain replicated. IFN-β production correlated with the inhibition of R. rickettsii Iowa. Reconstitution of Iowa with either of two recently described putative virulence determinants altered the IFN-β response. A rickettsial ankyrin repeat protein, RARP2, disrupts the trans-Golgi network and inhibits IFN-β secretion. An autotransporter peptidase, RapL, restores proteolytic maturation of outer membrane autotransporters and diminishes the IFN-β response to enhance cell survival and permit replication of the recombinant strain. These studies point the way toward discovery of mechanisms for innate immune response avoidance by virulent rickettsia.
Collapse
Affiliation(s)
- Liam Fitzsimmons
- Host-Parasite Interactions Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - DeAnna Bublitz
- Host-Parasite Interactions Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Tina Clark
- Host-Parasite Interactions Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Ted Hackstadt
- Host-Parasite Interactions Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| |
Collapse
|
14
|
Cheng R, Zhou C, Zhao M, Zhang S, Wan W, Yu Y, Wen B, Jiao J, Xiong X, Xu Q, OuYang X. TRIM56-mediated production of type I interferon inhibits intracellular replication of Rickettsia rickettsii. Microbiol Spectr 2024; 12:e0369523. [PMID: 38358243 PMCID: PMC10986528 DOI: 10.1128/spectrum.03695-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/18/2024] [Indexed: 02/16/2024] Open
Abstract
Rickettsia rickettsii (R. rickettsii), the causative agent of Rocky Mountain spotted fever (RMSF), is the most pathogenic member among Rickettsia spp. Previous studies have shown that tripartite motif-containing 56 (TRIM56) E3 ligase-induced ubiquitination of STING is important for cytosolic DNA sensing and type I interferon production to induce anti-DNA viral immunity, but whether it affects intracellular replication of R. rickettsii remains uncharacterized. Here, we investigated the effect of TRIM56 on HeLa and THP-1 cells infected with R. rickettsii. We found that the expression of TRIM56 was upregulated in the R. rickettsii-infected cells, and the overexpression of TRIM56 inhibited the intracellular replication of R. rickettsii, while R. rickettsii replication was enhanced in the TRIM56-silenced host cells with the reduced phosphorylation of IRF3 and STING and the increased production of interferon-β. In addition, the mutation of the TRIM56 E3 ligase catalytic site impairs the inhibitory function against R. rickettsii in HeLa cells. Altogether, our study discovers that TRIM56 is a host restriction factor of R. rickettsii by regulating the cGAS-STING-mediated signaling pathway. This study gives new evidence for the role of TRIM56 in the innate immune response against intracellular bacterial infection and provides new therapeutic targets for RMSF. IMPORTANCE Given that Rickettsia rickettsii (R. rickettsii) is the most pathogenic member within the Rickettsia genus and serves as the causative agent of Rocky Mountain spotted fever, there is a growing need to explore host targets. In this study, we examined the impact of host TRIM56 on R. rickettsii infection in HeLa and THP-1 cells. We observed a significant upregulation of TRIM56 expression in R. rickettsii-infected cells. Remarkably, the overexpression of TRIM56 inhibited the intracellular replication of R. rickettsii, while silencing TRIM56 enhanced bacterial replication accompanied by reduced phosphorylation of IRF3 and STING, along with increased interferon-β production. Notably, the mutation of the TRIM56's E3 ligase catalytic site did not impede R. rickettsii replication in HeLa cells. Collectively, our findings provide novel insights into the role of TRIM56 as a host restriction factor against R. rickettsii through the modulation of the cGAS-STING signaling pathway.
Collapse
Affiliation(s)
- Ruxi Cheng
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Chunyu Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Mingliang Zhao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Shan Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Weiqiang Wan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Yonghui Yu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Bohai Wen
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Jun Jiao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Xiaolu Xiong
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Qin Xu
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xuan OuYang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| |
Collapse
|
15
|
Sit B, Lamason RL. Pathogenic Rickettsia spp. as emerging models for bacterial biology. J Bacteriol 2024; 206:e0040423. [PMID: 38315013 PMCID: PMC10883807 DOI: 10.1128/jb.00404-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024] Open
Abstract
Our understanding of free-living bacterial models like Escherichia coli far outpaces that of obligate intracellular bacteria, which cannot be cultured axenically. All obligate intracellular bacteria are host-associated, and many cause serious human diseases. Their constant exposure to the distinct biochemical niche of the host has driven the evolution of numerous specialized bacteriological and genetic adaptations, as well as innovative molecular mechanisms of infection. Here, we review the history and use of pathogenic Rickettsia species, which cause an array of vector-borne vascular illnesses, as model systems to probe microbial biology. Although many challenges remain in our studies of these organisms, the rich pathogenic and biological diversity of Rickettsia spp. constitutes a unique backdrop to investigate how microbes survive and thrive in host and vector cells. We take a bacterial-focused perspective and highlight emerging insights that relate to new host-pathogen interactions, bacterial physiology, and evolution. The transformation of Rickettsia spp. from pathogens to models demonstrates how recalcitrant microbes may be leveraged in the lab to tap unmined bacterial diversity for new discoveries. Rickettsia spp. hold great promise as model systems not only to understand other obligate intracellular pathogens but also to discover new biology across and beyond bacteria.
Collapse
Affiliation(s)
- Brandon Sit
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Rebecca L. Lamason
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
16
|
Ponce Nájera E, Lozano Lazcano V, Ploneda González C, Montoya Hinojosa M, González Oropeza D. Case Report: Fatal Rickettsiosis in Pregnancy. Am J Trop Med Hyg 2024; 110:320-322. [PMID: 38190746 PMCID: PMC10859820 DOI: 10.4269/ajtmh.23-0419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 10/17/2023] [Indexed: 01/10/2024] Open
Abstract
Rocky Mountain spotted fever (RMSF) is a tick-borne infection caused by Rickettsia rickettsii. We present a series of two cases of pregnant patients who showed up at the emergency room of a hospital in Nuevo León, Mexico. Both patients lived in environments where R. rickettsii is endemic and they presented with several days of symptoms, including fever. Both patients developed a rash and had stillbirths during their hospital stay. Treatment with doxycycline was delayed, with fatal results in both patients. Diagnosis of RMSF was confirmed via polymerase chain reaction assay postmortem. The need to link epidemiological clues with clinical data is critical in the diagnosis and early treatment of RMSF to prevent maternal deaths.
Collapse
Affiliation(s)
- Eduardo Ponce Nájera
- Hospital Regional Materno Infantil del Estado de Nuevo León, Nuevo León, Mexico
- Escuela de Medicina y Ciencias de la Salud Tecnológico de Monterrey, Nuevo León, Mexico
| | - Valeria Lozano Lazcano
- Hospital Regional Materno Infantil del Estado de Nuevo León, Nuevo León, Mexico
- Escuela de Medicina y Ciencias de la Salud Tecnológico de Monterrey, Nuevo León, Mexico
| | - César Ploneda González
- Hospital Metropolitano “Dr. Bernardo Sepúlveda’’ del Estado de Nuevo León, Nuevo León, Mexico
| | | | - Diego González Oropeza
- Hospital Regional Materno Infantil del Estado de Nuevo León, Nuevo León, Mexico
- Escuela de Medicina y Ciencias de la Salud Tecnológico de Monterrey, Nuevo León, Mexico
| |
Collapse
|
17
|
Londoño AF, Farner JM, Dillon M, Grab DJ, Kim Y, Scorpio DG, Dumler JS. Benidipine impairs innate immunity converting sublethal to lethal infections in a murine model of spotted fever rickettsiosis. PLoS Negl Trop Dis 2024; 18:e0011993. [PMID: 38408129 PMCID: PMC10919851 DOI: 10.1371/journal.pntd.0011993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 03/07/2024] [Accepted: 02/13/2024] [Indexed: 02/28/2024] Open
Abstract
Spotted fever group rickettsiae are tick-borne obligate intracellular bacteria that infect microvascular endothelial cells. Humans and mammalian infection results in endothelial cell barrier dysfunction and increased vascular permeability. We previously demonstrated that treatment of Rickettsia parkeri-infected cells with the calcium channel blocker benidipine significantly delayed vascular barrier permeability. Thus, we hypothesized that benidipine, known to be safe and effective for other clinical processes, could reduce rickettsia-induced vascular permeability in vivo in an animal model of spotted fever rickettsiosis. Based on liver, lung and brain vascular FITC-dextran extravasation studies, benidipine did not reliably impact vascular permeability. However, it precipitated a deleterious effect on responses to control sublethal R. parkeri infection. Animals treated with benidipine alone had no clinical signs or changes in histopathology and splenic immune cell distributions. Benidipine-treated infected animals had marked increases in tissue and blood bacterial loads, more extensive inflammatory histopathologic injury, and changes in splenic architecture and immune cell distributions potentially reflecting diminished Ca2+ signaling, reduced innate immune cell activation, and loss of rickettsial propagation control. Impaired T cell activation by R. parkeri antigen in the presence of benidipine was confirmed in vitro with the use of NKT cell hybridomas. The unexpected findings stand in stark contrast to recent discussions of the benefits of calcium channel blockers for viral infections and chronic infectious or inflammatory diseases. A role for calcium channel blockers in exacerbation of human rickettsiosis and acute inflammatory infections should be evaluated by a retrospective review of patient's outcomes and medications.
Collapse
Affiliation(s)
- Andrés F Londoño
- Henry M. Jackson Foundation for Advancement of Military Medicine, Bethesda, Maryland, United States of America
- Department of Pathology, School of Medicine, Uniformed Services University, Bethesda, Maryland, United States of America
| | - Jennifer M Farner
- Henry M. Jackson Foundation for Advancement of Military Medicine, Bethesda, Maryland, United States of America
- Emerging Infectious Disease Graduate Program, School of Medicine, Uniformed Services University, Bethesda, Maryland, United States of America
| | - Marlon Dillon
- Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Dennis J Grab
- Department of Pathology, School of Medicine, Uniformed Services University, Bethesda, Maryland, United States of America
| | - Yuri Kim
- Henry M. Jackson Foundation for Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | - Diana G Scorpio
- Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - J Stephen Dumler
- Department of Pathology, School of Medicine, Uniformed Services University, Bethesda, Maryland, United States of America
| |
Collapse
|
18
|
Sahni A, Alsing J, Narra HP, Montini M, Zafar Y, Sahni SK. Endothelial Mechanistic Target of Rapamycin Activation with Different Strains of R. rickettsii: Possible Role in Rickettsial Pathogenesis. Microorganisms 2024; 12:296. [PMID: 38399700 PMCID: PMC10892065 DOI: 10.3390/microorganisms12020296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 01/18/2024] [Accepted: 01/28/2024] [Indexed: 02/25/2024] Open
Abstract
Rickettsia rickettsii is an obligate intracellular pathogen that primarily targets endothelial cells (ECs), leading to vascular inflammation and dysfunction. Mechanistic target of rapamycin (mTOR) regulates several cellular processes that directly affect host immune responses to bacterial pathogens. Here, we infected ECs with two R. rickettsii strains, avirulent (Iowa) and highly virulent Sheila Smith (SS) to identify differences in the kinetics and/or intensity of mTOR activation to establish a correlation between mTOR response and bacterial virulence. Endothelial mTOR activation with the highly virulent SS strain was significantly higher than with the avirulent Iowa strain. Similarly, there was increased LC3-II lipidation with the virulent SS strain compared with the avirulent Iowa strain of R. rickettsii. mTOR inhibitors rapamycin and Torin2 significantly increased bacterial growth and replication in the ECs, as evidenced by a more than six-fold increase in rickettsia copy numbers at 48 h post-infection. Further, the knockdown of mTOR with Raptor and Rictor siRNA resulted in a higher rickettsial copy number and the altered expression of the pro-inflammatory cytokines interleukin (IL)-1α, IL-6, and IL-8. These results are the first to reveal that endothelial mTOR activation and the early induction of autophagy might be governed by bacterial virulence and have established the mTOR pathway as an important regulator of endothelial inflammation, host immunity, and microbial replication.
Collapse
Affiliation(s)
- Abha Sahni
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555-0609, USA; (J.A.); (H.P.N.); (M.M.); (Y.Z.)
| | | | | | | | | | - Sanjeev K. Sahni
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555-0609, USA; (J.A.); (H.P.N.); (M.M.); (Y.Z.)
| |
Collapse
|
19
|
Dahmani M, Zhu JC, Cook JH, Riley SP. Anaphylatoxin signaling activates macrophages to control intracellular Rickettsia proliferation. Microbiol Spectr 2023; 11:e0253823. [PMID: 37855623 PMCID: PMC10714731 DOI: 10.1128/spectrum.02538-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/11/2023] [Indexed: 10/20/2023] Open
Abstract
IMPORTANCE Pathogenic Rickettsia species are extremely dangerous bacteria that grow within the cytoplasm of host mammalian cells. In most cases, these bacteria are able to overpower the host cell and grow within the protected environment of the cytoplasm. However, a dramatic conflict occurs when Rickettsia encounter innate immune cells; the bacteria can "win" by taking over the host, or the bacteria can "lose" if the host cell efficiently fights the infection. This manuscript examines how the immune complement system is able to detect the presence of Rickettsia and alert nearby cells. Byproducts of complement activation called anaphylatoxins are signals that "activate" innate immune cells to mount an aggressive defensive strategy. This study enhances our collective understanding of the innate immune reaction to intracellular bacteria and will contribute to future efforts at controlling these dangerous infections.
Collapse
Affiliation(s)
- Mustapha Dahmani
- Department of Veterinary Medicine, University of Maryland-College Park, College Park, Maryland, USA
| | - Jinyi C. Zhu
- Department of Veterinary Medicine, University of Maryland-College Park, College Park, Maryland, USA
| | - Jack H. Cook
- Department of Veterinary Medicine, University of Maryland-College Park, College Park, Maryland, USA
| | - Sean P. Riley
- Department of Veterinary Medicine, University of Maryland-College Park, College Park, Maryland, USA
- Virginia-Maryland College of Veterinary Medicine, College Park, Maryland, USA
| |
Collapse
|
20
|
Voss OH, Gaytan H, Ullah S, Sadik M, Moin I, Rahman MS, Azad AF. Autophagy facilitates intracellular survival of pathogenic rickettsiae in macrophages via evasion of autophagosomal maturation and reduction of microbicidal pro-inflammatory IL-1 cytokine responses. Microbiol Spectr 2023; 11:e0279123. [PMID: 37819111 PMCID: PMC10715094 DOI: 10.1128/spectrum.02791-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/01/2023] [Indexed: 10/13/2023] Open
Abstract
IMPORTANCE Rickettsia spp. are intracellular bacterial parasites of a wide range of arthropod and vertebrate hosts. Some rickettsiae are responsible for several severe human diseases globally. One interesting feature of these pathogens is their ability to exploit host cytosolic defense responses to their benefits. However, the precise mechanism by which pathogenic Rickettsia spp. elude host defense responses remains unclear. Here, we observed that pathogenic Rickettsia typhi and Rickettsia rickettsii (Sheila Smith [SS]), but not non-pathogenic Rickettsia montanensis, become ubiquitinated and induce autophagy upon entry into macrophages. Moreover, unlike R. montanensis, R. typhi and R. rickettsii (SS) colocalized with LC3B but not with Lamp2 upon host cell entry. Finally, we observed that both R. typhi and R. rickettsii (SS), but not R. montanensis, reduce pro-inflammatory interleukin-1 (IL-1) responses, likely via an autophagy-mediated mechanism. In summary, we identified a previously unappreciated pathway by which both pathogenic R. typhi and R. rickettsii (SS) become ubiquitinated, induce autophagy, avoid autolysosomal destruction, and reduce microbicidal IL-1 cytokine responses to establish an intracytosolic niche in macrophages.
Collapse
Affiliation(s)
- Oliver H. Voss
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Hodalis Gaytan
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Saif Ullah
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Mohammad Sadik
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Imran Moin
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - M. Sayeedur Rahman
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Abdu F. Azad
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
21
|
Chavarría-Bencomo IV, Nevárez-Moorillón GV, Espino-Solís GP, Adame-Gallegos JR. Antibiotic resistance in tick-borne bacteria: A One Health approach perspective. J Infect Public Health 2023; 16 Suppl 1:153-162. [PMID: 37945496 DOI: 10.1016/j.jiph.2023.10.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 11/12/2023] Open
Abstract
The emergence and re-emergence of tick-borne bacteria (TBB) as a public health problem raises the uncertainty of antibiotic resistance in these pathogens, which could be dispersed to other pathogens. The impact of global warming has led to the emergence of pathogenic TBB in areas where they were not previously present and is another risk that must be taken into account under the One Health guides. This review aimed to analyze the existing information regarding antibiotic-resistant TBB and antibiotic-resistance genes (ARG) present in the tick microbiome, considering the potential to be transmitted to pathogenic microorganisms. Several Ehrlichia species have been reported to exhibit natural resistance to fluoroquinolones and typhus group Rickettsiae are naturally susceptible to erythromycin. TBB have a lower risk of acquiring ARG due to their natural habitat, but there is still a probability of acquiring them; furthermore, studies of these pathogens are limited. Pathogenic and commensal bacteria coexist within the tick microbiome along with ARGs for antibiotic deactivation, cellular protection, and efflux pumps; these ARGs confer resistance to antibiotics such as aminoglycosides, beta-lactamase, diaminopyrimidines, fluoroquinolones, glycopeptides, sulfonamides, and tetracyclines. Although with low probability, TBB can be a reservoir of ARGs.
Collapse
Affiliation(s)
- Inés Valeria Chavarría-Bencomo
- Facultad de Ciencias Químicas. Universidad Autónoma de Chihuahua, Circuito Universitario s/n. Campus Universitario II., 31125 Chihuahua, Mexico
| | - Guadalupe Virginia Nevárez-Moorillón
- Facultad de Ciencias Químicas. Universidad Autónoma de Chihuahua, Circuito Universitario s/n. Campus Universitario II., 31125 Chihuahua, Mexico.
| | - Gerardo Pavel Espino-Solís
- Laboratorio Nacional de Citometría de Flujo. Facultad de Medicina y Ciencias Biomédicas, Universidad Autónoma de Chihuahua, Circuito Universitario s/n. Campus Universitario II., 31125 Chihuahua, Mexico
| | - Jaime Raúl Adame-Gallegos
- Facultad de Ciencias Químicas. Universidad Autónoma de Chihuahua, Circuito Universitario s/n. Campus Universitario II., 31125 Chihuahua, Mexico
| |
Collapse
|
22
|
Zhang Y, Hai Y, Duan B, Long H, Xie X, Teng Z, Yin F, Wang M, Xiong Y, Shao Z, Guo W, Qin A. A seminested recombinase polymerase amplification assay to detect rickettsial pathogens in clinical samples. Diagn Microbiol Infect Dis 2023; 107:116067. [PMID: 37751629 DOI: 10.1016/j.diagmicrobio.2023.116067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/14/2023] [Accepted: 08/18/2023] [Indexed: 09/28/2023]
Abstract
Treatment at the early stage of onset is vital for the prognosis of rickettsioses. But the absence of specific clinical symptoms complicates the diagnosis of this condition. Herein we established a seminested recombinase polymerase amplification assay (snRPA-nfo) that enables quick detection and differentiation of rickettsial pathogens in clinical samples with high sensitivity and specificity. The conserved 17-kDa protein gene of Rickettsia sibirica and the 47-kDa protein gene of Orientia tsutsugamushi were targeted for the duplex RPA-nfo assay. The snRPA-nfo assay exhibited an increased LOD in spiked blood samples, up to 1000-fold in comparison to standard RPA-nfo, and a better detection rate (83.3%, 5/6) than TaqMan PCR (16.6%, 1/6, Ct ≤ 35) in clinically confirmed patient blood samples. Thus, snRPA-nfo assay represents a promising alternative to TaqMan PCR in the early diagnosis of rickettsioses for point-of-care testing as well as in resource-limited settings.
Collapse
Affiliation(s)
- Ying Zhang
- Center for Disease Control and Prevention of Xilingol League, Xilinhaote, Inner Mongolia, China; State Key Laboratory of Infectious Diseases Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yan Hai
- Center for Disease Control and Prevention of Inner Mongolia, Hohhot, Inner Mongolia, China
| | - Biao Duan
- Institute of Endemic Diseases Control and Prevention of Yunnan, Dali, Yunnan, China
| | - Hu Long
- Center for Disease Control and Prevention of Guilin City, Guilin, Guangxi, China
| | - Xiaofei Xie
- State Key Laboratory of Infectious Diseases Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China; Hainan Medical College, Haikou, Hainan, China
| | - Zhongqiu Teng
- State Key Laboratory of Infectious Diseases Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Feifei Yin
- Hainan Medical College, Haikou, Hainan, China
| | - Mingliu Wang
- Center for Disease Control and Prevention of Guangxi, Nanning, Guangxi, China
| | - Yanwen Xiong
- State Key Laboratory of Infectious Diseases Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zhujun Shao
- State Key Laboratory of Infectious Diseases Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Weidong Guo
- Center for Disease Control and Prevention of Xilingol League, Xilinhaote, Inner Mongolia, China; Institute of Endemic Diseases Control and Prevention of Yunnan, Dali, Yunnan, China.
| | - Aiping Qin
- State Key Laboratory of Infectious Diseases Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China.
| |
Collapse
|
23
|
Bei J, Qiu Y, Cockrell D, Chang Q, Husseinzadeh S, Zhou C, Fang X, Bao X, Jin Y, Gaitas A, Khanipov K, Saito TB, Gong B. Identification of common sequence motifs shared exclusively among selectively packed exosomal pathogenic microRNAs during rickettsial infections. J Cell Physiol 2023; 238:1937-1948. [PMID: 37334929 DOI: 10.1002/jcp.31061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/12/2023] [Accepted: 05/22/2023] [Indexed: 06/21/2023]
Abstract
We previously reported that microRNA (miR)23a and miR30b are selectively sorted into exosomes derived from rickettsia-infected endothelial cells (R-ECExos). Yet, the mechanism remains unknown. Cases of spotted fever rickettsioses have been increasing, and infections with these bacteria cause life-threatening diseases by targeting brain and lung tissues. Therefore, the goal of the present study is to further dissect the molecular mechanism underlying R-ECExos-induced barrier dysfunction of normal recipient microvascular endothelial cells (MECs), depending on their exosomal RNA cargos. Infected ticks transmit the rickettsiae to human hosts following a bite and injections of the bacteria into the skin. In the present study, we demonstrate that treatment with R-ECExos, which were derived from spotted fever group R parkeri infected human dermal MECs, induced disruptions of the paracellular adherens junctional protein VE-cadherin, and breached the paracellular barrier function in recipient pulmonary MECs (PMECs) in an exosomal RNA-dependent manner. We did not detect different levels of miRs in parent dermal MECs following rickettsial infections. However, we demonstrated that the microvasculopathy-relevant miR23a-27a-24 cluster and miR30b are selectively enriched in R-ECExos. Bioinformatic analysis revealed that common sequence motifs are shared exclusively among the exosomal, selectively-enriched miR23a cluster and miR30b at different levels. Taken together, these data warrant further functional identification and characterization of a monopartition, bipartition, or tripartition among ACA, UCA, and CAG motifs that guide recognition of microvasculopathy-relevant miR23a-27a-24 and miR30b, and subsequently results in their selective enrichments in R-ECExos.
Collapse
Affiliation(s)
- Jiani Bei
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Yuan Qiu
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Diane Cockrell
- Laboratory of Bacteriology, Division of Intramural Research, NIAID-NIH, Hamilton, Montana, USA
| | - Qing Chang
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Sorosh Husseinzadeh
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Changcheng Zhou
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Xiang Fang
- Department of Neurology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Xiaoyong Bao
- Department of Pediatric, University of Texas Medical Branch, Galveston, Texas, USA
| | - Yang Jin
- Department of Medicine, Pulmonary and Critical Care Medicine Division, Boston University Medical Campus, Boston, Massachusetts, USA
| | - Angelo Gaitas
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kamil Khanipov
- Department of Pharmacology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Tais B Saito
- Laboratory of Bacteriology, Division of Intramural Research, NIAID-NIH, Hamilton, Montana, USA
| | - Bin Gong
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
24
|
Kim Y, Clemens EG, Farner JM, Londono-Barbaran A, Grab DJ, Dumler JS. Spotted fever rickettsia-induced microvascular endothelial barrier dysfunction is delayed by the calcium channel blocker benidipine. Biochem Biophys Res Commun 2023; 663:96-103. [PMID: 37121130 PMCID: PMC10362780 DOI: 10.1016/j.bbrc.2023.04.045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 04/17/2023] [Indexed: 05/02/2023]
Abstract
The tick-borne bacterium Rickettsia parkeri is an obligate intracellular pathogen that belongs to spotted fever group rickettsia (SFGR). The SFG pathogens are characterized by their ability to infect and rapidly proliferate inside host vascular endothelial cells that eventually result in impairment of vascular endothelium barrier functions. Benidipine, a wide range dihydropyridine calcium channel blocker, is used to prevent and treat cardiovascular diseases. In this study, we tested whether benidipine has protective effects against rickettsia-induced microvascular endothelial cell barrier dysfunction in vitro. We utilized an in vitro vascular model consisting of transformed human brain microvascular endothelial cells (tHBMECs) and continuously monitored transendothelial electric resistance (TEER) across the cell monolayer. We found that during the late stages of infection when we observed TEER decrease and when there was a gradual increase of the cytoplasmic [Ca2+], benidipine prevented these rickettsia-induced effects. In contrast, nifedipine, another cardiovascular dihydropyridine channel blocker specific for L-type Ca2+ channels, did not prevent R. parkeri-induced drop of TEER. Additionally, neither drug was bactericidal. These data suggest that growth of R. parkeri inside endothelial cells is associated with impairment of endothelial cell monolayer integrity due to Ca2+ flooding through specific, benidipine-sensitive T- or N/Q-type Ca2+ channels but not through nifedipine-sensitive L-type Ca2+ channels. Further study will be required to discern the exact nature of the Ca2+ channels and Ca2+ transporting system(s) involved, any contributions of the pathogen toward this process, as well as the suitability of benidipine and new dihydropyridine derivatives as complimentary therapeutic drugs against Rickettsia-induced vascular failure.
Collapse
Affiliation(s)
- Yuri Kim
- Henry M. Jackson Foundation for the Advancement of Military Medicine, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA; Uniformed Services of the Health Sciences, Department of Pathology, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| | - Emily G Clemens
- Uniformed Services of the Health Sciences, Department of Pathology, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| | - Jennifer M Farner
- Henry M. Jackson Foundation for the Advancement of Military Medicine, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA; Uniformed Services of the Health Sciences, Department of Pathology, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| | - Andres Londono-Barbaran
- Henry M. Jackson Foundation for the Advancement of Military Medicine, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA; Uniformed Services of the Health Sciences, Department of Pathology, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| | - Dennis J Grab
- Uniformed Services of the Health Sciences, Department of Pathology, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| | - J Stephen Dumler
- Uniformed Services of the Health Sciences, Department of Pathology, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| |
Collapse
|
25
|
Londoño AF, Scorpio DG, Dumler JS. Innate immunity in rickettsial infections. Front Cell Infect Microbiol 2023; 13:1187267. [PMID: 37228668 PMCID: PMC10203653 DOI: 10.3389/fcimb.2023.1187267] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 04/19/2023] [Indexed: 05/27/2023] Open
Abstract
Rickettsial agents are a diverse group of alpha-proteobacteria within the order Rickettsiales, which possesses two families with human pathogens, Rickettsiaceae and Anaplasmataceae. These obligate intracellular bacteria are most frequently transmitted by arthropod vectors, a first step in the pathogens' avoidance of host cell defenses. Considerable study of the immune responses to infection and those that result in protective immunity have been conducted. Less study has focused on the initial events and mechanism by which these bacteria avoid the innate immune responses of the hosts to survive within and propagate from host cells. By evaluating the major mechanisms of evading innate immunity, a range of similarities among these bacteria become apparent, including mechanisms to escape initial destruction in phagolysosomes of professional phagocytes, those that dampen the responses of innate immune cells or subvert signaling and recognition pathways related to apoptosis, autophagy, proinflammatory responses, and mechanisms by which these microbes attach to and enter cells or those molecules that trigger the host responses. To illustrate these principles, this review will focus on two common rickettsial agents that occur globally, Rickettsia species and Anaplasma phagocytophilum.
Collapse
Affiliation(s)
- Andrés F. Londoño
- The Henry M. Jackson Foundation for Advancement in Military Medicine, Bethesda, MD, United States
- Department of Pathology, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Diana G. Scorpio
- Host-Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - J. Stephen Dumler
- Department of Pathology, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
26
|
Cente M, Danchenko M, Skultety L, Filipcik P, Sekeyova Z. Rickettsia Deregulates Genes Coding for the Neurotoxic Cell Response Pathways in Cerebrocortical Neurons In Vitro. Cells 2023; 12:cells12091235. [PMID: 37174635 PMCID: PMC10177168 DOI: 10.3390/cells12091235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Rickettsial infections of the central nervous system (CNS) are manifested by severe neurological symptoms and represent a serious life-threatening condition. Despite the considerable health danger, only a few studies have been conducted focusing on the pathogenesis induced by Rickettsia sp. in CNS. To investigate the signaling pathways associated with the neurotoxic effects of rickettsiae, we employed an experimental model of cerebrocortical neurons combined with molecular profiling and comprehensive bioinformatic analysis. The cytopathic effect induced by Rickettsia akari and Rickettsia slovaca was demonstrated by decreased neuronal viability, structural changes in cell morphology, and extensive fragmentation of neurites in vitro. Targeted profiling revealed the deregulation of genes involved in the neuroinflammatory and neurotoxic cell response pathways. Although quantitative analysis showed differences in gene expression response, functional annotation revealed that the biological processes are largely shared between both Rickettsia species. The identified enriched pathways are associated with cytokine signaling, chemotaxis of immune cells, responses to infectious agents, interactions between neurons, endothelial and glial cells, and regulation of neuronal apoptotic processes. The findings of our study provide new insight into the etiopathogenesis of CNS infection and further expand the understanding of molecular signaling associated with neuroinvasive Rickettsia species.
Collapse
Affiliation(s)
- Martin Cente
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10 Bratislava, Slovakia
| | - Monika Danchenko
- Department of Rickettsiology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Ludovit Skultety
- Department of Rickettsiology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Peter Filipcik
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10 Bratislava, Slovakia
| | - Zuzana Sekeyova
- Department of Rickettsiology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| |
Collapse
|
27
|
Du LF, Zhang MZ, Yuan TT, Ni XB, Wei W, Cui XM, Wang N, Xiong T, Zhang J, Pan YS, Zhu DY, Li LJ, Xia LY, Wang TH, Wei R, Liu HB, Sun Y, Zhao L, Lam TTY, Cao WC, Jia N. New insights into the impact of microbiome on horizontal and vertical transmission of a tick-borne pathogen. MICROBIOME 2023; 11:50. [PMID: 36915209 PMCID: PMC10012463 DOI: 10.1186/s40168-023-01485-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 02/01/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND The impact of host skin microbiome on horizontal transmission of tick-borne pathogens , and of pathogen associated transstadial and transovarial changes in tick microbiome are largely unknown, but are important to control increasingly emerging tick-borne diseases worldwide. METHODS Focusing on a rickettsiosis pathogen, Rickettsia raoultii, we used R. raoultii-positive and R. raoultii-negative Dermacentor spp. tick colonies to study the involvement of skin microbiota in cutaneous infection with rickettsiae in laboratory mice, and the function of the tick microbiome on maintenance of rickettsiae through all tick developmental stages (eggs, larvae, nymphs, adults) over two generations. RESULTS We observed changes in the skin bacteria community, such as Chlamydia, not only associated with rickettsial colonization but also with tick feeding on skin. The diversity of skin microbiome differed between paired tick-bitten and un-bitten sites. For vertical transmission, significant differences in the tick microbiota between pathogenic rickettsia-positive and -negative tick chorts was observed across all developmental stages at least over two generations, which appeared to be a common pattern not only for R. raoultii but also for another pathogenic species, Candidatus Rickettsia tarasevichiae. More importantly, bacterial differences were complemented by functional shifts primed for genetic information processing during blood feeding. Specifically, the differences in tick microbiome gene repertoire between pathogenic Rickettsia-positive and -negative progenies were enriched in pathways associated with metabolism and hormone signals during vertical transmission. CONCLUSIONS We demonstrate that host skin microbiome might be a new factor determining the transmission of rickettsial pathogens through ticks. While pathogenic rickettsiae infect vertebrate hosts during blood-feeding by the tick, they may also manipulate the maturation of the tick through changing the functional potential of its microbiota over the tick's life stages. The findings here might spur the development of new-generation control methods for ticks and tick-borne pathogens. Video Abstract.
Collapse
Affiliation(s)
- Li-Feng Du
- Institute of EcoHealth, School of Public Health, Shandong University, 44 Wenhuaxi Street, Jinan, 250012, Shandong, People's Republic of China
| | - Ming-Zhu Zhang
- Institute of EcoHealth, School of Public Health, Shandong University, 44 Wenhuaxi Street, Jinan, 250012, Shandong, People's Republic of China
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, People's Republic of China
| | - Ting-Ting Yuan
- School of Medicine, Nankai University, Tianjin, 300071, People's Republic of China
| | - Xue-Bing Ni
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, The University of Hong Kong, Hong Kong SAR, People's Republic of China
- Laboratory of Data Discovery for Health Limited, 19W Hong Kong Science & Technology Parks, Hong Kong SAR, People's Republic of China
| | - Wei Wei
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, People's Republic of China
| | - Xiao-Ming Cui
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, People's Republic of China
| | - Ning Wang
- Institute of EcoHealth, School of Public Health, Shandong University, 44 Wenhuaxi Street, Jinan, 250012, Shandong, People's Republic of China
| | - Tao Xiong
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, People's Republic of China
| | - Jie Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, People's Republic of China
| | - Yu-Sheng Pan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, People's Republic of China
| | - Dai-Yun Zhu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, People's Republic of China
| | - Liang-Jing Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, People's Republic of China
| | - Luo-Yuan Xia
- Institute of EcoHealth, School of Public Health, Shandong University, 44 Wenhuaxi Street, Jinan, 250012, Shandong, People's Republic of China
| | - Tian-Hong Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, People's Republic of China
| | - Ran Wei
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, People's Republic of China
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, People's Republic of China
| | - Hong-Bo Liu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, People's Republic of China
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, People's Republic of China
| | - Yi Sun
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, People's Republic of China
| | - Lin Zhao
- Institute of EcoHealth, School of Public Health, Shandong University, 44 Wenhuaxi Street, Jinan, 250012, Shandong, People's Republic of China
| | - Tommy Tsan-Yuk Lam
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, The University of Hong Kong, Hong Kong SAR, People's Republic of China.
- Laboratory of Data Discovery for Health Limited, 19W Hong Kong Science & Technology Parks, Hong Kong SAR, People's Republic of China.
- Guangdong-Hongkong Joint Laboratory of Emerging Infectious Diseases, Joint Institute of Virology (Shantou University/The University of Hong Kong), Shantou, Guangdong, 515063, People's Republic of China.
- EKIH (Gewuzhikang) Pathogen Research Institute, Futian District, Shenzhen City, Guangdong, 518045, People's Republic of China.
- Centre for Immunology & Infection Limited, 17W Hong Kong Science & Technology Parks, Hong Kong SAR, People's Republic of China.
| | - Wu-Chun Cao
- Institute of EcoHealth, School of Public Health, Shandong University, 44 Wenhuaxi Street, Jinan, 250012, Shandong, People's Republic of China.
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, People's Republic of China.
| | - Na Jia
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, People's Republic of China.
| |
Collapse
|
28
|
Verhoeve VI, Lehman SS, Driscoll TP, Beckmann JF, Gillespie JJ. Metagenome diversity illuminates origins of pathogen effectors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.26.530123. [PMID: 36909625 PMCID: PMC10002696 DOI: 10.1101/2023.02.26.530123] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
Recent metagenome assembled genome (MAG) analyses have profoundly impacted Rickettsiology systematics. Discovery of basal lineages (Mitibacteraceae and Athabascaceae) with predicted extracellular lifestyles reveals an evolutionary timepoint for the transition to host dependency, which occurred independent of mitochondrial evolution. Notably, these basal rickettsiae carry the Rickettsiales vir homolog (rvh) type IV secretion system (T4SS) and purportedly use rvh to kill congener microbes rather than parasitize host cells as described for derived rickettsial pathogens. MAG analysis also substantially increased diversity for genus Rickettsia and delineated a basal lineage (Tisiphia) that stands to inform on the rise of human pathogens from protist and invertebrate endosymbionts. Herein, we probed Rickettsiales MAG and genomic diversity for the distribution of Rickettsia rvh effectors to ascertain their origins. A sparse distribution of most Rickettsia rvh effectors outside of Rickettsiaceae lineages indicates unique rvh evolution from basal extracellular species and other rickettsial families. Remarkably, nearly every effector was found in multiple divergent forms with variable architectures, illuminating profound roles for gene duplication and recombination in shaping effector repertoires in Rickettsia pathogens. Lateral gene transfer plays a prominent role shaping the rvh effector landscape, as evinced by the discover of many effectors on plasmids and conjugative transposons, as well as pervasive effector gene exchange between Rickettsia and Legionella species. Our study exemplifies how MAGs can provide incredible insight on the origins of pathogen effectors and how their architectural modifications become tailored to eukaryotic host cell biology.
Collapse
Affiliation(s)
- Victoria I Verhoeve
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Stephanie S Lehman
- Division of Molecular and Cellular Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Timothy P Driscoll
- Department of Biology, West Virginia University, Morgantown, West Virginia, USA
| | - John F Beckmann
- Microbiology and Immunology, University of South Alabama, Mobile, AL, USA
| | - Joseph J Gillespie
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
29
|
Bei J, Qiu Y, Cockrell D, Chang Q, Husseinzadeh S, Zhou C, Gaitas A, Fang X, Jin Y, Khanipov K, Saito TB, Gong B. Identification of common sequence motifs shared exclusively among selectively packed exosomal pathogenic microRNAs during rickettsial infections. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.06.522907. [PMID: 36712112 PMCID: PMC9881928 DOI: 10.1101/2023.01.06.522907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
We previously reported that microRNA (miR)23a and miR30b are selectively sorted into rickettsia-infected, endothelial cell-derived exosomes ( R -ECExos). Yet, the mechanism remains unknown. The number of cases of spotted fever rickettsioses has been increasing in recent years, and infections with these bacteria cause life-threatening diseases by targeting brain and lung tissues. Therefore, the aim of the present study is to continue to dissect the molecular mechanism underlying R -ECExos-induced barrier dysfunction of normal recipient microvascular endothelial cells (MECs), depending on their exosomal RNA cargos. Rickettsiae are transmitted to human hosts by the bite of an infected tick into the skin. In the present study we demonstrate that treatment with R -ECExos, which were derived from spotted fever group R parkeri infected human dermal MECs, induced disruptions of the paracellular adherens junctional protein VE-cadherin and breached the paracellular barrier function in recipient pulmonary MECs (PMECs) in an exosomal RNA-dependent manner. Similarly, we did not detect different levels of miRs in parent dermal MECs following rickettsial infections. However, we demonstrated that the microvasculopathy-relevant miR23a-27a-24 cluster and miR30b are selectively enriched in R -ECExos. Bioinformatic analysis revealed that common sequence motifs are shared exclusively among the exosomal, selectively-enriched miR23a cluster and miR30b at different levels. Taken together, these data warrant further functional identification and characterization of a single, bipartition, or tripartition among ACA, UCA, and CAG motifs that guide recognition of microvasculopathy-relevant miR23a-27a-24 and miR30b, and subsequently results in their selective enrichments in R -ECExos.
Collapse
|
30
|
Tripathi AK, Ray AK, Mishra SK, Bishen SM, Mishra H, Khurana A. Molecular and Therapeutic Insights of Alpha-Lipoic Acid as a Potential Molecule for Disease Prevention. REVISTA BRASILEIRA DE FARMACOGNOSIA : ORGAO OFICIAL DA SOCIEDADE BRASILEIRA DE FARMACOGNOSIA 2023; 33:272-287. [PMID: 36778891 PMCID: PMC9904877 DOI: 10.1007/s43450-023-00370-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 01/25/2023] [Indexed: 02/10/2023]
Abstract
Alpha-lipoic acid is an organic, sulfate-based compound produced by plants, humans, and animals. As a potent antioxidant and a natural dithiol compound, it performs a crucial role in mitochondrial bioenergetic reactions. A healthy human body, on the other hand, can synthesize enough α-lipoic acid to scavenge reactive oxygen species and increase endogenous antioxidants; however, the amount of α-lipoic acid inside the body decreases significantly with age, resulting in endothelial dysfunction. Molecular orbital energy and spin density analysis indicate that the sulfhydryl (-SH) group of molecules has the greatest electron donating activity, which would be responsible for the antioxidant potential and free radical scavenging activity. α-Lipoic acid acts as a chelating agent for metal ions, a quenching agent for reactive oxygen species, and a reducing agent for the oxidized form of glutathione and vitamins C and E. α-Lipoic acid enantiomers and its reduced form have antioxidant, cognitive, cardiovascular, detoxifying, anti-aging, dietary supplement, anti-cancer, neuroprotective, antimicrobial, and anti-inflammatory properties. α-Lipoic acid has cytotoxic and antiproliferative effects on several cancers, including polycystic ovarian syndrome. It also has usefulness in the context of female and male infertility. Although α-lipoic acid has numerous clinical applications, the majority of them stem from its antioxidant properties; however, its bioavailability in its pure form is low (approximately 30%). However, nanoformulations have shown promise in this regard. The proton affinity and electron donating activity, as a redox-active agent, would be responsible for the antioxidant potential and free radical scavenging activity of the molecule. This review discusses the most recent clinical data on α-lipoic acid in the prevention, management, and treatment of a variety of diseases, including coronavirus disease 2019. Based on current evidence, the preclinical and clinical potential of this molecule is discussed. Graphical Abstract Supplementary Information The online version contains supplementary material available at 10.1007/s43450-023-00370-1.
Collapse
Affiliation(s)
- Amit Kumar Tripathi
- School of Basic and Applied Science, Galgotias University, Gautam Buddha Nagar, UP Noida, India
- Molecular Biology Unit, Institute of Medical Science, Banaras Hindu University, Varanasi, 221005 India
| | - Anup Kumar Ray
- Department of Natural Products, National Institute of Pharmaceutical Education and Research, Sahibzada Ajit Singh Nagar, Punjab, India
| | - Sunil Kumar Mishra
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, 221005 India
| | - Siddharth Mall Bishen
- Department of Physics, Banaras Hindu University, Mahila Maha Vidyalaya, Varanasi, India
| | - Hirdyesh Mishra
- Department of Physics, Banaras Hindu University, Mahila Maha Vidyalaya, Varanasi, India
| | - Aman Khurana
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, 221005 India
| |
Collapse
|
31
|
Gao S, Li L, Zhou X, Dai X, Lu L, Chen Y, Han J, Huang X, Xiao Q, He H, Liu Q. Fatal Rickettsia Japonica Infection Complicating Disseminated Intravascular Coagulation in Yichang, China. Infect Drug Resist 2022; 15:6613-6623. [PMID: 36386421 PMCID: PMC9664911 DOI: 10.2147/idr.s383917] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 10/18/2022] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND Severe complications may cause a fatal or disabling outcome in patients with Rickettsia japonica infection but are poorly understood. METHODS We identified 11 patients with only Rickettsia japonica infection with metagenomics next generation sequencing (mNGS) during April to November 2021 at Yichang Central People's Hospital, China. Clinical data were obtained through review of medical records. RESULTS Most patients realized that they had symptoms about one or two days after being bitten. Fever (91%), pulmonary effusion (91%), rash or erythema (100%), abnormal urine (100%), neutropenia (100%), lymphopenia (100%), and thrombocytopenia (100%) were the most common clinical signs. Six severely ill patients were admitted to the intensive care unit and five had mild symptoms. Systemic manifestations such as vomiting (83%), neurological manifestations (100%), and disseminated intravascular coagulation (100%) were more frequently observed in the severe cases, 33.3% of whom developed purpura fulminans requiring amputation or skin graft, and 16.6% died two days after admission. Some patients experienced sequelae. CONCLUSION Our study found that patients with critical Rickettsia japonica infection complicating disseminated intravascular coagulation had high risk of poor outcome.
Collapse
Affiliation(s)
- Siyu Gao
- Yichang Central People’s Hospital, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, People’s Republic of China
| | - Lingfeng Li
- Department of Critical Care Medicine, Dongguan Institute of Respiratory and Critical Care Medicine, Dongguan People’s Hospital, Dongguan, Guangdong, People’s Republic of China
| | - Xiaolin Zhou
- Yichang Central People’s Hospital, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, People’s Republic of China
| | - Xiangcheng Dai
- Yichang Central People’s Hospital, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, People’s Republic of China
| | - Lu Lu
- Yichang Central People’s Hospital, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, People’s Republic of China
| | - Yandong Chen
- Yichang Central People’s Hospital, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, People’s Republic of China
| | - Jing Han
- Yichang Central People’s Hospital, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, People’s Republic of China
| | - Xiaoli Huang
- Yichang Central People’s Hospital, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, People’s Republic of China
| | - Qian Xiao
- Yichang Central People’s Hospital, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, People’s Republic of China
| | - Huiqing He
- Centers for Disease Control and Prevention, Yichang, Hubei, People’s Republic of China
| | - Qiang Liu
- Yichang Central People’s Hospital, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, People’s Republic of China
| |
Collapse
|
32
|
Huang D, Luo J, OuYang X, Song L. Subversion of host cell signaling: The arsenal of Rickettsial species. Front Cell Infect Microbiol 2022; 12:995933. [PMID: 36389139 PMCID: PMC9659576 DOI: 10.3389/fcimb.2022.995933] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 10/04/2022] [Indexed: 10/10/2023] Open
Abstract
Rickettsia is a genus of nonmotile, Gram-negative, non-spore-forming, highly pleomorphic bacteria that cause severe epidemic rickettsioses. The spotted fever group and typhi group are major members of the genus Rickettsia. Rickettsial species from the two groups subvert diverse host cellular processes, including membrane dynamics, actin cytoskeleton dynamics, phosphoinositide metabolism, intracellular trafficking, and immune defense, to promote their host colonization and intercellular transmission through secreted effectors (virulence factors). However, lineage-specific rickettsiae have exploited divergent strategies to accomplish such challenging tasks and these elaborated strategies focus on distinct host cell processes. In the present review, we summarized current understandings of how different rickettsial species employ their effectors' arsenal to affect host cellular processes in order to promote their own replication or to avoid destruction.
Collapse
Affiliation(s)
- Dan Huang
- Department of Respiratory Medicine, Center of Pathogen Biology and Infectious Disease, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Jingjing Luo
- Department of Respiratory Medicine, Center of Pathogen Biology and Infectious Disease, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Xuan OuYang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Lei Song
- Department of Respiratory Medicine, Center of Pathogen Biology and Infectious Disease, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
33
|
Sethi SK, Dhaliwal M, Raghunathan V, Raaj H, Mahato SK, Vadhera A, Dragon-Durey MA, Jha PK, Bansal SB, Raina R. Siblings with complement activation, thrombotic microangiopathy and gangrene: Answers. Pediatr Nephrol 2022; 37:2057-2058. [PMID: 35166915 DOI: 10.1007/s00467-022-05464-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/04/2022] [Indexed: 10/19/2022]
Affiliation(s)
- Sidharth Kumar Sethi
- Pediatric Nephrology, Kidney Institute, Medanta, The Medicity, Gurgaon, Haryana, 122001, India
| | - Maninder Dhaliwal
- Pediatric Critical Care, Medanta, The Medicity, Gurgaon, Haryana, India, 122001
| | - Veena Raghunathan
- Pediatric Critical Care, Medanta, The Medicity, Gurgaon, Haryana, India, 122001
| | - Hritik Raaj
- Maulana Azad Medical College, New Delhi, 110002, India
| | | | | | | | - Pranaw Kumar Jha
- Kidney Institute, Medanta, The Medicity, Gurgaon, Haryana, 122001, India
| | | | - Rupesh Raina
- Pediatric Nephrology, Akron Children's Hospital, Akron, OH, USA.
| |
Collapse
|
34
|
Abstract
Ticks are hematophagous ectoparasites capable of transmitting multiple human pathogens. Environmental changes have supported the expansion of ticks into new geographical areas that have become the epicenters of tick-borne diseases (TBDs). The spotted fever group (SFG) of Rickettsia frequently infects ticks and causes tick-transmitted rickettsioses in areas of endemicity where ixodid ticks support host transmission during blood feeding. Ticks also serve as a reservoir for SFG Rickettsia. Among the members of SFG Rickettsia, R. rickettsii causes Rocky Mountain spotted fever (RMSF), the most lethal TBD in the United States. Cases of RMSF have been reported for over a century in association with several species of ticks in the United States. However, the isolation of R. rickettsii from ticks has decreased, and recent serological and epidemiological studies suggest that novel species of SFG Rickettsia are responsible for the increased number of cases of RMSF-like rickettsioses in the United States. Recent analyses of rickettsial genomes and advances in genetic and molecular studies of Rickettsia provided insights into the biology of Rickettsia with the identification of conserved and unique putative virulence genes involved in the rickettsial life cycle. Thus, understanding Rickettsia-host-tick interactions mediating successful disease transmission and pathogenesis for SFG rickettsiae remains an active area of research. This review summarizes recent advances in understanding how SFG Rickettsia species coopt and manipulate ticks and mammalian hosts to cause rickettsioses, with a particular emphasis on newly described or emerging SFG Rickettsia species.
Collapse
|
35
|
Involvement of Pore Formation and Osmotic Lysis in the Rapid Killing of Gamma Interferon-Pretreated C166 Endothelial Cells by Rickettsia prowazekii. Trop Med Infect Dis 2022; 7:tropicalmed7080163. [PMID: 36006255 PMCID: PMC9415803 DOI: 10.3390/tropicalmed7080163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 07/24/2022] [Accepted: 07/25/2022] [Indexed: 11/16/2022] Open
Abstract
Rickettsia prowazekii, the bacterial cause of epidemic typhus in humans, proliferates mainly within the microvascular endothelial cells. Previous studies have shown that murine macrophage-like RAW264.7 cells are rapidly damaged if they are pretreated with gamma interferon (IFN-γ) and then infected with R. prowazekii. In the present study, the effects of IFN-γ and R. prowazekii on murine C166 endothelial cells were evaluated. In the IFN-γ-pretreated R. prowazekii-infected endothelial cell cultures, evidence of cell damage was observed within several hours after addition of the rickettsiae. Considerable numbers of the cells became permeable to trypan blue dye and ethidium bromide, and substantial amounts of lactate dehydrogenase (LDH) were released from the cells. Such evidence of cellular injury was not observed in the untreated infected cultures or in any of the mock-infected cultures. Polyethylene glycols (PEGs) of different nominal average molecular weights were used to assess the possible involvement of pore formation and osmotic lysis in this cellular injury. PEG 8000 dramatically suppressed LDH release, PEG 4000 partially inhibited it, and PEGs 2000 and 1450 had no effect. Despite its inhibition of LDH release, PEG 8000 did not prevent the staining of the IFN-γ-pretreated infected endothelial cells by ethidium bromide. These findings suggest that the observed cellular injury involves the formation of pores in the endothelial cell membranes, followed by osmotic lysis of the cells.
Collapse
|
36
|
Groth M, Skrzydlewska E, Dobrzyńska M, Pancewicz S, Moniuszko-Malinowska A. Redox Imbalance and Its Metabolic Consequences in Tick-Borne Diseases. Front Cell Infect Microbiol 2022; 12:870398. [PMID: 35937690 PMCID: PMC9353526 DOI: 10.3389/fcimb.2022.870398] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 06/13/2022] [Indexed: 11/21/2022] Open
Abstract
One of the growing global health problems are vector-borne diseases, including tick-borne diseases. The most common tick-borne diseases include Lyme disease, tick-borne encephalitis, human granulocytic anaplasmosis, and babesiosis. Taking into account the metabolic effects in the patient's body, tick-borne diseases are a significant problem from an epidemiological and clinical point of view. Inflammation and oxidative stress are key elements in the pathogenesis of infectious diseases, including tick-borne diseases. In consequence, this leads to oxidative modifications of the structure and function of phospholipids and proteins and results in qualitative and quantitative changes at the level of lipid mediators arising in both reactive oxygen species (ROS) and ROS enzyme-dependent reactions. These types of metabolic modifications affect the functioning of the cells and the host organism. Therefore, links between the severity of the disease state and redox imbalance and the level of phospholipid metabolites are being searched, hoping to find unambiguous diagnostic biomarkers. Assessment of molecular effects of oxidative stress may also enable the monitoring of the disease process and treatment efficacy.
Collapse
Affiliation(s)
- Monika Groth
- Department of Infectious Diseases and Neuroinfections, Medical University of Bialystok, Bialystok, Poland
| | - Elżbieta Skrzydlewska
- Department of Inorganic and Analytical Chemistry, Medical University of Bialystok, Bialystok, Poland
| | - Marta Dobrzyńska
- Department of Inorganic and Analytical Chemistry, Medical University of Bialystok, Bialystok, Poland
| | - Sławomir Pancewicz
- Department of Infectious Diseases and Neuroinfections, Medical University of Bialystok, Bialystok, Poland
| | - Anna Moniuszko-Malinowska
- Department of Infectious Diseases and Neuroinfections, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
37
|
Zhou C, Bei J, Qiu Y, Chang Q, Nyong E, Vasilakis N, Yang J, Krishnan B, Khanipov K, Jin Y, Fang X, Gaitas A, Gong B. Exosomally Targeting microRNA23a Ameliorates Microvascular Endothelial Barrier Dysfunction Following Rickettsial Infection. Front Immunol 2022; 13:904679. [PMID: 35812423 PMCID: PMC9260018 DOI: 10.3389/fimmu.2022.904679] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/23/2022] [Indexed: 11/25/2022] Open
Abstract
Spotted fever group rickettsioses caused by Rickettsia (R) are devastating human infections, which mainly target microvascular endothelial cells (ECs) and can induce lethal EC barrier dysfunction in the brain and lungs. Our previous evidence reveals that exosomes (Exos) derived from rickettsial-infected ECs, namely R-ECExos, can induce disruption of the tight junctional (TJ) protein ZO-1 and barrier dysfunction of human normal recipient brain microvascular endothelial cells (BMECs). However, the underlying mechanism remains elusive. Given that we have observed that microRNA23a (miR23a), a negative regulator of endothelial ZO-1 mRNA, is selectively sorted into R-ECExos, the aim of the present study was to characterize the potential functional role of exosomal miR23a delivered by R-ECExos in normal recipient BMECs. We demonstrated that EC-derived Exos (ECExos) have the capacity to deliver oligonucleotide RNAs to normal recipient BMECs in an RNase-abundant environment. miR23a in ECExos impairs normal recipient BMEC barrier function, directly targeting TJ protein ZO-1 mRNAs. In separate studies using a traditional in vitro model and a novel single living-cell biomechanical assay, our group demonstrated that miR23a anti-sense oligonucleotide-enriched ECExos ameliorate R-ECExo-provoked recipient BMEC dysfunction in association with stabilization of ZO-1 in a dose-dependent manner. These results suggest that Exo-based therapy could potentially prove to be a promising strategy to improve vascular barrier function during bacterial infection and concomitant inflammation.
Collapse
Affiliation(s)
- Changcheng Zhou
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Jiani Bei
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Yuan Qiu
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Qing Chang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Emmanuel Nyong
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Nikos Vasilakis
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
- Department of Preventive Medicine and Population Health, The University of Texas Medical Branch, Galveston, TX, United States
- Center for Vector Borne and Zoonotic Diseases, University of Texas Medical Branch, Galveston, TX, United States
- The Center of Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, United States
- Center for Tropical Diseases, University of Texas Medical Branch, Galveston, TX, United States
- Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, TX, United States
| | - Jun Yang
- Department of Internal Medicine, Endocrinology, University of Texas Medical Branch, Galveston, TX, United States
| | - Balaji Krishnan
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch, Galveston, TX, United States
| | - Kamil Khanipov
- Department of Pharmacology, University of Texas Medical Branch, Galveston, TX, United States
| | - Yang Jin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University Medical Campus, Boston, MA, United States
| | - Xiang Fang
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch, Galveston, TX, United States
| | - Angelo Gaitas
- The Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Bin Gong
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
- Center for Vector Borne and Zoonotic Diseases, University of Texas Medical Branch, Galveston, TX, United States
- The Center of Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, United States
- Center for Tropical Diseases, University of Texas Medical Branch, Galveston, TX, United States
- Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
38
|
Brown Dog Tick ( Rhipicephalus sanguineus Sensu Lato) Infection with Endosymbiont and Human Pathogenic Rickettsia spp., in Northeastern México. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19106249. [PMID: 35627785 PMCID: PMC9141927 DOI: 10.3390/ijerph19106249] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/17/2022] [Accepted: 05/17/2022] [Indexed: 11/23/2022]
Abstract
Of the documented tick-borne diseases infecting humans in México, Rocky Mountain spotted fever (RMSF), caused by the Gram-negative bacterium Rickettsia rickettsii, is responsible for most fatalities. Given recent evidence of brown dog tick, Rhipicephalus sanguineus s.l., as an emerging vector of human RMSF, we aimed to evaluate dogs and their ticks for rickettsiae infections as an initial step in assessing the establishment of this pathosystem in a poorly studied region of northeastern México while evaluating the use of dogs as sentinels for transmission/human disease risk. We sampled owned dogs living in six disadvantaged neighborhoods of Reynosa, northeastern México to collect whole blood and ticks. Of 168 dogs assessed, tick infestation prevalence was 53%, composed of exclusively Rh. sanguineus s. l. (n = 2170 ticks). Using PCR and sequencing, we identified an overall rickettsiae infection prevalence of 4.1% (n = 12/292) in ticks, in which eight dogs harbored at least one infected tick. Rickettsiae infections included Rickettsia amblyommatis and Rickettsia parkeri, both of which are emerging human pathogens, as well as Candidatus Rickettsia andeanae. This is the first documentation of pathogenic Rickettsia species in Rh. sanguineus s.l. collected from dogs from northeastern México. Domestic dog infestation with Rickettsia-infected ticks indicates ongoing transmission; thus, humans are at risk for exposure, and this underscores the importance of public and veterinary health surveillance for these pathogens.
Collapse
|
39
|
Pillay A, Manyangadze T, Mukaratirwa S. Prevalence of Rickettsia africae in tick vectors collected from mammalian hosts in sub-Saharan Africa: A systematic review and meta-analysis. Ticks Tick Borne Dis 2022; 13:101960. [DOI: 10.1016/j.ttbdis.2022.101960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 04/08/2022] [Accepted: 04/21/2022] [Indexed: 11/29/2022]
|
40
|
Sensing the Messenger: Potential Roles of Cyclic-di-GMP in Rickettsial Pathogenesis. Int J Mol Sci 2022; 23:ijms23073853. [PMID: 35409212 PMCID: PMC8999164 DOI: 10.3390/ijms23073853] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/28/2022] [Accepted: 03/30/2022] [Indexed: 02/04/2023] Open
Abstract
Pathogenic bacteria causing human rickettsioses, transmitted in nature by arthropod vectors, primarily infect vascular endothelial cells lining the blood vessels, resulting in 'endothelial activation' and onset of innate immune responses. Nucleotide second messengers are long presumed to be the stimulators of type I interferons, of which bacterial cyclic-di-GMP (c-di-GMP) has been implicated in multiple signaling pathways governing communication with other bacteria and host cells, yet its importance in the context of rickettsial interactions with the host has not been investigated. Here, we report that all rickettsial genomes encode a putative diguanylate cyclase pleD, responsible for the synthesis of c-di-GMP. In silico analysis suggests that although the domain architecture of PleD is apparently well-conserved among different rickettsiae, the protein composition and sequences likely vary. Interestingly, cloning and sequencing of the pleD gene from virulent (Sheila Smith) and avirulent (Iowa) strains of R. rickettsii reveals a nonsynonymous substitution, resulting in an amino acid change (methionine to isoleucine) at position 236. Additionally, a previously reported 5-bp insertion in the genomic sequence coding for pleD (NCBI accession: NC_009882) was not present in the sequence of our cloned pleD from R. rickettsii strain Sheila Smith. In vitro infection of HMECs with R. rickettsii (Sheila Smith), but not R. rickettsii (Iowa), resulted in dynamic changes in the levels of pleD up to 24 h post-infection. These findings thus provide the first evidence for the potentially important role(s) of c-di-GMP in the determination of host-cell responses to pathogenic rickettsiae. Further studies into molecular mechanisms through which rickettsial c-di-GMP might regulate pathogen virulence and host responses should uncover the contributions of this versatile bacterial second messenger in disease pathogenesis and immunity to human rickettsioses.
Collapse
|
41
|
Belkhir S, Hamede R, Thomas F, Ujvari B, Dujon AM. Season, weight, and age, but not transmissible cancer, affect tick loads in the endangered Tasmanian devil. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2022; 98:105221. [PMID: 35065301 DOI: 10.1016/j.meegid.2022.105221] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/12/2022] [Accepted: 01/17/2022] [Indexed: 11/28/2022]
Abstract
The Tasmanian devil (Sarcophilus harrisii) is a carnivorous marsupial threatened by a transmissible cancer, devil facial tumour disease (DFTD). While we have a good understanding of the effect of the transmissible cancer on its host, little information is available about its potential interactions with ectoparasites. With this study, we aimed to determine the factors driving tick loads in a DFTD affected Tasmanian devil population, using long-term mark-recapture data. We investigated the effect of a range of life history traits (age, weight, sex, body condition) and of DFTD (time since DFTD arrival and presence of tumours) on the ectoparasitic tick load of the devils. Mixed effect models revealed that tick load in Tasmanian devils was primarily driven by season, weight, body condition and age. Young devils had more ticks compared to older or healthier devils. The reduction in Tasmanian devil population size over the past 14 years at the studied site had little effect on tick infestation. We also found that devils infected by DFTD had a similar tick load compared to those free of observable tumours, suggesting no interaction between the transmissible cancer and tick load. Our study highlights seasonality and life cycle as primary drivers of tick infestation in Tasmanian devils and the need for further investigations to integrate devil stress and immune dynamics with ectoparasite counts.
Collapse
Affiliation(s)
- Sophia Belkhir
- Deakin University, Geelong, School of Life and Environmental Sciences, Centre for Integrative Ecology, Waurn Ponds, Vic, 3216, Australia; École normale supérieure de Lyon, Département de biologie, Master biologie, Lyon, France
| | - Rodrigo Hamede
- School of Natural Sciences, University of Tasmania, Private Bag 55, Hobart, Tasmania 7001, Australia
| | - Frédéric Thomas
- CREEC, MIVEGEC, UMR IRD 224-CNRS 5290-Université de Montpellier, Montpellier, France
| | - Beata Ujvari
- Deakin University, Geelong, School of Life and Environmental Sciences, Centre for Integrative Ecology, Waurn Ponds, Vic, 3216, Australia
| | - Antoine M Dujon
- Deakin University, Geelong, School of Life and Environmental Sciences, Centre for Integrative Ecology, Waurn Ponds, Vic, 3216, Australia; CREEC, MIVEGEC, UMR IRD 224-CNRS 5290-Université de Montpellier, Montpellier, France.
| |
Collapse
|
42
|
Matos AL, Curto P, Simões I. Moonlighting in Rickettsiales: Expanding Virulence Landscape. Trop Med Infect Dis 2022; 7:32. [PMID: 35202227 PMCID: PMC8877226 DOI: 10.3390/tropicalmed7020032] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/05/2022] [Accepted: 02/17/2022] [Indexed: 12/22/2022] Open
Abstract
The order Rickettsiales includes species that cause a range of human diseases such as human granulocytic anaplasmosis (Anaplasma phagocytophilum), human monocytic ehrlichiosis (Ehrlichia chaffeensis), scrub typhus (Orientia tsutsugamushi), epidemic typhus (Rickettsia prowazekii), murine typhus (R. typhi), Mediterranean spotted fever (R. conorii), or Rocky Mountain spotted fever (R. rickettsii). These diseases are gaining a new momentum given their resurgence patterns and geographical expansion due to the overall rise in temperature and other human-induced pressure, thereby remaining a major public health concern. As obligate intracellular bacteria, Rickettsiales are characterized by their small genome sizes due to reductive evolution. Many pathogens employ moonlighting/multitasking proteins as virulence factors to interfere with multiple cellular processes, in different compartments, at different times during infection, augmenting their virulence. The utilization of this multitasking phenomenon by Rickettsiales as a strategy to maximize the use of their reduced protein repertoire is an emerging theme. Here, we provide an overview of the role of various moonlighting proteins in the pathogenicity of these species. Despite the challenges that lie ahead to determine the multiple potential faces of every single protein in Rickettsiales, the available examples anticipate this multifunctionality as an essential and intrinsic feature of these obligates and should be integrated into available moonlighting repositories.
Collapse
Affiliation(s)
- Ana Luísa Matos
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (A.L.M.); (P.C.)
| | - Pedro Curto
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (A.L.M.); (P.C.)
| | - Isaura Simões
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (A.L.M.); (P.C.)
- IIIUC—Institute of Interdisciplinary Research, University of Coimbra, 3004-504 Coimbra, Portugal
| |
Collapse
|
43
|
Biswas S, Ghosh R, Roy D, Ray A, De K, Biswas S, Naga D, Benito-León J. Scrub Typhus Masquerading as Limbic Encephalitis. Neurohospitalist 2022; 12:105-110. [PMID: 34950396 PMCID: PMC8689554 DOI: 10.1177/19418744211016107] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Scrub typhus, an acute febrile infectious disease prevalent in the 'tsutsugamushi triangle', is a mite-born rickettsial zoonosis, caused by Orientia tsutsugamushi. The clinical presentation is protean and involves multiple organ systems of the body, including central and peripheral nervous systems. We report a 22-year-old previously healthy Indian woman who presented with clinical (confusion, excessive sleepiness, cognitive dysfunction and focal seizures) and neuroimaging features of limbic encephalitis. After exclusion of common infectious, autoimmune and paraneoplastic causes, she was diagnosed with scrub typhus associated encephalitis, which responded to doxycycline and azithromycin therapy.
Collapse
Affiliation(s)
- Subhrajyoti Biswas
- Department of General Medicine, Burdwan Medical College, Burdwan, West Bengal, India
| | - Ritwik Ghosh
- Department of General Medicine, Burdwan Medical College, Burdwan, West Bengal, India
| | - Dipayan Roy
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Jodhpur, Rajasthan, India,Indian Institute of Technology (IIT), Madras, Tamil Nadu, India
| | - Adrija Ray
- Department of General Medicine, RG Kar Medical College & Hospital, Kolkata, West Bengal, India
| | - Kaustav De
- Department of General Medicine, Burdwan Medical College, Burdwan, West Bengal, India
| | - Subrata Biswas
- Department of General Medicine, Burdwan Medical College, Burdwan, West Bengal, India
| | - Dinabandhu Naga
- Department of General Medicine, Burdwan Medical College, Burdwan, West Bengal, India
| | - Julián Benito-León
- Department of Neurology, University Hospital “12 de Octubre”, Madrid, Spain,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain,Department of Medicine, Complutense University, Madrid, Spain,Julián Benito-León. Department of Neurology, University Hospital “12 de Octubre”, 28041 Madrid, Spain.
| |
Collapse
|
44
|
Pacheco-Silva AB, Martins EB, López AJR, Detepo PJT, Mamani RF, Japiassú AM, Lupi O, Mendes TV, Pina-Costa AD, Calvet GA, Bressan CDS, Silva MFBD, Ogino LL, Assis MRDS, Marques AM, Dias CMG, Borchert JM, Lemos ERSD, Brasil P. Fatal Brazilian spotted fever in a healthy military man during field training in Rio de Janeiro city, southeastern Brazil. Rev Inst Med Trop Sao Paulo 2022; 64:e77. [DOI: 10.1590/s1678-9946202264077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/20/2022] [Indexed: 11/16/2022] Open
|
45
|
Spotted Fever Group Rickettsia Trigger Species-Specific Alterations in Macrophage Proteome Signatures with Different Impacts in Host Innate Inflammatory Responses. Microbiol Spectr 2021; 9:e0081421. [PMID: 34935429 PMCID: PMC8693926 DOI: 10.1128/spectrum.00814-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The molecular details underlying differences in pathogenicity between Rickettsia species remain to be fully understood. Evidence points to macrophage permissiveness as a key mechanism in rickettsial virulence. Different studies have shown that several rickettsial species responsible for mild forms of rickettsioses can also escape macrophage-mediated killing mechanisms and establish a replicative niche within these cells. However, their manipulative capacity with respect to host cellular processes is far from being understood. A deeper understanding of the interplay between mildly pathogenic rickettsiae and macrophages and the commonalities and specificities of host responses to infection would illuminate differences in immune evasion mechanisms and pathogenicity. We used quantitative proteomics by sequential windowed data independent acquisition of the total high-resolution mass spectra with tandem mass spectrometry (SWATH-MS/MS) to profile alterations resulting from infection of THP-1 macrophages with three mildly pathogenic rickettsiae: Rickettsia parkeri, Rickettsia africae, and Rickettsia massiliae, all successfully proliferating in these cells. We show that all three species trigger different proteome signatures. Our results reveal a significant impact of infection on proteins categorized as type I interferon responses, which here included several components of the retinoic acid-inducible gene I (RIG-1)-like signaling pathway, mRNA splicing, and protein translation. Moreover, significant differences in protein content between infection conditions provide evidence for species-specific induced alterations. Indeed, we confirm distinct impacts on host inflammatory responses between species during infection, demonstrating that these species trigger different levels of beta interferon (IFN-β), differences in the bioavailability of the proinflammatory cytokine interleukin 1β (IL-1β), and differences in triggering of pyroptotic events. This work reveals novel aspects and exciting nuances of macrophage-Rickettsia interactions, adding additional layers of complexity between Rickettsia and host cells' constant arms race for survival. IMPORTANCE The incidence of diseases caused by Rickettsia has been increasing over the years. It has long been known that rickettsioses comprise diseases with a continuous spectrum of severity. There are highly pathogenic species causing diseases that are life threatening if untreated, others causing mild forms of the disease, and a third group for which no pathogenicity to humans has been described. These marked differences likely reflect distinct capacities for manipulation of host cell processes, with macrophage permissiveness emerging as a key virulence trait. However, what defines pathogenicity attributes among rickettsial species is far from being resolved. We demonstrate that the mildly pathogenic Rickettsia parkeri, Rickettsia africae, and Rickettsia massiliae, all successfully proliferating in macrophages, trigger different proteome signatures in these cells and differentially impact critical components of innate immune responses by inducing different levels of beta interferon (IFN-β) and interleukin 1β (IL-1β) and different timing of pyroptotic events during infection. Our work reveals novel nuances in rickettsia-macrophage interactions, offering new clues to understand Rickettsia pathogenicity.
Collapse
|
46
|
Dahmani M, Cook JH, Zhu JC, Riley SP. Contribution of classical complement activation and IgM to the control of Rickettsia infection. Mol Microbiol 2021; 116:1476-1488. [PMID: 34725868 PMCID: PMC8955150 DOI: 10.1111/mmi.14839] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 01/02/2023]
Abstract
Pathogenic Rickettsia are obligate intracellular bacteria and the etiologic agents of many life‐threatening infectious diseases. Due to the serious nature of these infections, it is imperative to both identify the responsive immune sensory pathways and understand the associated immune mechanisms that restrict Rickettsia proliferation. Previous studies have demonstrated that the mammalian complement system is both activated during Rickettsia infection and contributes to the immune response to infection. To further define this component of the mammalian anti‐Rickettsia immune response, we sought to identify the mechanism(s) of complement activation during Rickettsia infection. We have employed a series of in vitro and in vivo models of infection to investigate the role of the classical complement activation pathway during Rickettsia infection. Depletion or elimination of complement activity demonstrates that both C1q and pre‐existing IgM contribute to complement activation; thus implicating the classical complement system in Rickettsia‐mediated complement activation. Elimination of the classical complement pathway from mice increases susceptibility to R. australis infection with both increased bacterial loads in multiple tissues and decreased immune activation markers. This study highlights the role of the classical complement pathway in immunity against Rickettsia and implicates resident Rickettsia‐responsive IgM in the response to infection.
Collapse
Affiliation(s)
- Mustapha Dahmani
- Department of Veterinary Medicine, University of Maryland-College Park, College Park, Maryland, USA.,Virginia-Maryland College of Veterinary Medicine, College Park, Maryland, USA
| | - Jack H Cook
- Department of Veterinary Medicine, University of Maryland-College Park, College Park, Maryland, USA.,Virginia-Maryland College of Veterinary Medicine, College Park, Maryland, USA
| | - Jinyi C Zhu
- Department of Veterinary Medicine, University of Maryland-College Park, College Park, Maryland, USA.,Virginia-Maryland College of Veterinary Medicine, College Park, Maryland, USA
| | - Sean P Riley
- Department of Veterinary Medicine, University of Maryland-College Park, College Park, Maryland, USA.,Virginia-Maryland College of Veterinary Medicine, College Park, Maryland, USA
| |
Collapse
|
47
|
Uribe Pulido N, Escorcia García C, Cabrera Orrego R, Gutiérrez LA, Agudelo CA. Acute Q Fever With Dermatologic Manifestations, Molecular Diagnosis, and No Seroconversion. Open Forum Infect Dis 2021; 8:ofab458. [PMID: 34692888 PMCID: PMC8530256 DOI: 10.1093/ofid/ofab458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 09/06/2021] [Indexed: 11/14/2022] Open
Abstract
We herein described a case of acute infection by Coxiella burnetii (acute Q fever) that started with a short incubation period and showed prominent dermatological manifestations and unusual serological behavior. The infection was confirmed by molecular detection through real-time polymerase chain reaction using genomic DNA collected from peripheral blood.
Collapse
Affiliation(s)
| | | | - Ruth Cabrera Orrego
- Grupo Biología de Sistemas, Escuela de Ciencias de la Salud, Facultad de Medicina, Universidad Pontificia Bolivariana, Medellín, Colombia
| | - Lina Andrea Gutiérrez
- Grupo Biología de Sistemas, Escuela de Ciencias de la Salud, Facultad de Medicina, Universidad Pontificia Bolivariana, Medellín, Colombia
| | - Carlos Andrés Agudelo
- Clínica Universitaria Bolivariana, Medellín, Colombia
- Escuela de Ciencias de la Salud, Facultad de Medicina, Universidad Pontificia Bolivariana, Medellín, Colombia
| |
Collapse
|
48
|
Blumenthal R, Hoosen A, Skosana LB, Weyer J. A Fatal Case of Rickettsiosis From South Africa. Am J Forensic Med Pathol 2021; 42:e57-e58. [PMID: 33788777 DOI: 10.1097/paf.0000000000000671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
ABSTRACT We present a case of fatal Rickettsiosis from South Africa.
Collapse
Affiliation(s)
| | - Anwar Hoosen
- Medical Microbiology, University of Pretoria, Pretoria
| | | | | |
Collapse
|
49
|
Burke TP, Engström P, Tran CJ, Langohr IM, Glasner DR, Espinosa DA, Harris E, Welch MD. Interferon receptor-deficient mice are susceptible to eschar-associated rickettsiosis. eLife 2021; 10:e67029. [PMID: 34423779 PMCID: PMC8428839 DOI: 10.7554/elife.67029] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 07/31/2021] [Indexed: 01/08/2023] Open
Abstract
Arthropod-borne rickettsial pathogens cause mild and severe human disease worldwide. The tick-borne pathogen Rickettsia parkeri elicits skin lesions (eschars) and disseminated disease in humans; however, inbred mice are generally resistant to infection. We report that intradermal infection of mice lacking both interferon receptors (Ifnar1-/-;Ifngr1-/-) with as few as 10 R. parkeri elicits eschar formation and disseminated, lethal disease. Similar to human infection, eschars exhibited necrosis and inflammation, with bacteria primarily found in leukocytes. Using this model, we find that the actin-based motility factor Sca2 is required for dissemination from the skin to internal organs, and the outer membrane protein OmpB contributes to eschar formation. Immunizing Ifnar1-/-;Ifngr1-/- mice with sca2 and ompB mutant R. parkeri protects against rechallenge, revealing live-attenuated vaccine candidates. Thus, Ifnar1-/-;Ifngr1-/- mice are a tractable model to investigate rickettsiosis, virulence factors, and immunity. Our results further suggest that discrepancies between mouse and human susceptibility may be due to differences in interferon signaling.
Collapse
Affiliation(s)
- Thomas P Burke
- Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Patrik Engström
- Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Cuong J Tran
- Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
- Division of Infectious Disease and Vaccinology, School of Public Health, University of California, BerkeleyBerkeleyUnited States
| | - Ingeborg M Langohr
- Department of Pathobiological Sciences, Louisiana State UniversityBaton RougeUnited States
| | - Dustin R Glasner
- Division of Infectious Disease and Vaccinology, School of Public Health, University of California, BerkeleyBerkeleyUnited States
| | - Diego A Espinosa
- Division of Infectious Disease and Vaccinology, School of Public Health, University of California, BerkeleyBerkeleyUnited States
| | - Eva Harris
- Division of Infectious Disease and Vaccinology, School of Public Health, University of California, BerkeleyBerkeleyUnited States
| | - Matthew D Welch
- Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| |
Collapse
|
50
|
Abstract
Spotted fever group rickettsioses (SFRs) are devastating human infections. Vascular endothelial cells (ECs) are the primary targets of rickettsial infection. Edema resulting from EC barrier dysfunction occurs in the brain and lungs in most cases of lethal SFR, but the underlying mechanisms remain unclear. The aim of the study was to explore the potential role of Rickettsia-infected, EC-derived exosomes (Exos) during infection. Using size exclusion chromatography (SEC), we purified Exos from conditioned, filtered, bacterium-free media collected from Rickettsia parkeri-infected human umbilical vein ECs (HUVECs) (R-ECExos) and plasma of Rickettsia australis- or R. parkeri-infected mice (R-plsExos). We observed that rickettsial infection increased the release of heterogeneous plsExos, but endothelial exosomal size, morphology, and production were not significantly altered following infection. Compared to normal plsExos and ECExos, both R-plsExos and R-ECExos induced dysfunction of recipient normal brain microvascular ECs (BMECs). The effect of R-plsExos on mouse recipient BMEC barrier function is dose dependent. The effect of R-ECExos on human recipient BMEC barrier function is dependent on the exosomal RNA cargo. Next-generation sequencing analysis and stem-loop quantitative reverse transcription-PCR (RT-qPCR) validation revealed that rickettsial infection triggered the selective enrichment of endothelial exosomal mir-23a and mir-30b, which potentially target the endothelial barrier. To our knowledge, this is the first report on the functional role of extracellular vesicles following infection by obligately intracellular bacteria.
Collapse
|